DOI:
10.1039/D4LC00877D
(Critical Review)
Lab Chip, 2025,
25, 1128-1148
Droplet microfluidics: unveiling the hidden complexity of the human microbiome
Received
16th October 2024
, Accepted 17th December 2024
First published on 8th January 2025
Abstract
The human body harbors diverse microbial communities essential for maintaining health and influencing disease processes. Droplet microfluidics, a precise and high-throughput platform for manipulating microscale droplets, has become vital in advancing microbiome research. This review introduces the foundational principles of droplet microfluidics, its operational capabilities, and wide-ranging applications. We emphasize its role in enhancing single-cell sequencing technologies, particularly genome and RNA sequencing, transforming our understanding of microbial diversity, gene expression, and community dynamics. We explore its critical function in isolating and cultivating traditionally unculturable microbes and investigating microbial activity and interactions, facilitating deeper insight into community behavior and metabolic functions. Lastly, we highlight its broader applications in microbial analysis and its potential to revolutionize human health research by driving innovations in diagnostics, therapeutic development, and personalized medicine. This review provides a comprehensive overview of droplet microfluidics' impact on microbiome research, underscoring its potential to transform our understanding of microbial dynamics and their relevance to health and disease.
1. Introduction
The human microbiome regulates various physiological functions, including digestion, metabolism, and immune response.1,2 Approximately 10–100 trillion multi-kingdom microbes, primarily bacteria but also archaea, protozoa, fungi, and viruses, inhabit the human body.3 These microbes contain 50 to 100 times more genetic information than the human host. Recent advances in multi-omics technologies, such as high-throughput sequencing, along with large-scale initiatives such as the integrative human microbiome project (iHMP) and the metagenomics of the human intestinal tract (MetaHIT), have significantly expanded our understanding of microbial composition and community structure at populational levels. Moreover, researchers have leveraged bioinformatics tools, including metagenomic analysis, metabolic network analysis, and machine learning, to explore associations between specific microbial taxa and host health outcomes. For instance, with the help of causality validation through microbial isolation, cultivation, and animal modeling, numerous studies have proved how probiotics can improve host immunity and regulate metabolic pathways while pathogenic microbes invade tissues and cause diseases.4
Despite significant advances, several challenges continue to impede microbiome research. One major obstacle is the interindividual variability in microbiome composition, which complicates efforts to generalize findings and predict clinical outcomes across diverse populations. Furthermore, microbial communities' spatiotemporal composition and gene expression are influenced by diet,5 immune status,6 and circadian rhythms.7 Another challenge lies in the substantial genomic variation between strains within the same species,8,9 which can significantly affect their metabolic potential, antibiotic resistance, and roles in host immunity and pathogenicity. Finally, although metagenomic studies have revealed the importance of unculturable microbes,10 the lack of isolation and validation remains a critical barrier to fully understanding their functions. To address these issues, developing high-throughput cultivation methods and in vitro models that mimic the host environment is crucial for uncovering these microbes' specific effects and mechanisms.
Microfluidics, the applied manipulation of fluid at the microscale, offers distinct and complementary solutions to many challenges in microbiome research.11 In particular, droplet microfluidics process picoliter to nanoliter aqueous droplets within an immiscible carrier fluid,12 such as fluorinated oil, with each droplet as a miniaturized reaction chamber. This technology enables the isolation of individual microbial cells, providing a platform for high-throughput, dynamic analysis of microbial physiology and genetic material. It is precious for addressing interindividual variability by allowing single-cell resolved analysis and capturing dynamic interactions between microbes and their environment. Moreover, droplet microfluidics facilitates the cultivation and functional screening of previously unculturable microbes, bridging the gap between correlation and causality in microbiome studies. Integrating droplet microfluidics with advanced sequencing and analytical tools offers unprecedented insights into microbial interactions.
This review will explore the diverse applications of droplet microfluidics in microbiome research, focusing on microbial single-cell sequencing, cultivation, functional screening, and interaction studies (Scheme 1). We will highlight how these technologies reshape our understanding of microbiome dynamics and their impact on host health. Special attention will be given to how droplet microfluidics is used to overcome challenges in isolating unculturable microbes, enabling high-throughput functional assays, and advancing microbial community synthesis and targeted delivery. This review aims to provide a comprehensive overview of how droplet microfluidics advances microbiome research and paves the way for new therapeutic approaches.
 |
| Scheme 1 Applications of droplet microfluidics in human microbiome research, highlighting fundamental techniques and their diverse applications. | |
2. The complexity and diversity of human microbiota
The complexity and diversity of the human microbiota have been extensively reviewed.13,14 Here, we provide a brief overview of the physiological and pathological aspects of the human microbiome. Microbial colonization occurs at birth, or even in utero, with over 50% of an infant's microbiota originating from maternal sources, including nasopharynx, saliva, skin, breast milk, feces, and the vaginal microbiota.15 Consequently, the infant microbial population initially mirrors the maternal microbiota during the first weeks or months, gradually evolving to a more complex structure by 12 months and resembling the adult microbiome by age three.16 Numerous factors influence this colonization process, including genetics, body environments, breastfeeding, diet, medication, and geographic location. These factors contribute to human microbiota's remarkable intra- and inter-individual diversity.
Recent studies have attempted to categorize the human microbiome into core and variable components to better understand the relationship between dysbiosis and health conditions.17,18 Multi-kingdom microbiota has been applied as biomarkers for disease diagnosis and prognosis, such as colorectal cancer19 and autism spectrum disorder.20 The core microbiome is primarily defined by microbial composition,21 though functional properties,22 ecological parameters,23 and temporal stability24 also play significant roles. Here, Table 1 highlights the predominant microbes identified across various body sites through sequencing and their associations with dysbiosis-related diseases, emphasizing the critical role of the microbiome in human health.
Table 1 Predominant multi-kingdom microbiota across different body sites and key dysbiosis-associated species
Sites |
Predominant multi-kingdom microbes |
Dysbiosis-related diseases and core species |
Bacteriome |
Mycobiome25,26 |
Archaeome27,28 |
Virome29 |
Gut2,18,21,30 |
Firmicutes |
Saccharomyces, Malassezia, Candida |
Methanobrevibacter
|
Bacteriophage |
Inflammatory bowel diseases (IBD)31–33 |
Klebsiella pneumoniae
|
Bacteroidetes |
Clostridioides difficile
|
Proteobacteria |
Insulin resistance34 |
Candida albicans
|
Actinobacteria |
Colorectal cancer35 |
Increased Lachnospiraceae (family-level) |
Fusobacterium nucleatum
|
Respiratory tract36–39 |
Streptococcus, Veillonella, Prevotella, Haemophilus |
Candida, Saccharomyces, Penicillium |
Nanoarchaeota |
Bacteriophage, Anelloviridae, Redondoviridae |
Chronic obstructive pulmonary disease40 |
Haemophilus influenzae, Moraxella catarrhalis, Streptococcus pneumoniae |
Pulmonary fibrosis41,42 |
Increased Streptococcus, SARS-CoV-2 |
Chronic pulmonary aspergillosis43 |
Aspergillus species (commonly Aspergillus fumigatus) |
Tuberculosis44 |
Mycobacterium tuberculosis
|
Oral cavity39,45,46 |
Firmicutes |
Candida
|
Methanobrevibacter
|
Redondoviridae, Anelloviridae, Bacteriophages |
Periodontitis47 |
Porphyromonas gingivalis
|
Actinobacteria |
Dental caries48 |
Candida albicans
|
Proteobacteria |
Apical periodontitis49,50 |
Streptococcus mutans
|
Fusobacteria |
Enterococcus faecalis
|
Bacteroidetes |
Spirochaetes |
Skin51,52 |
Actinobacteria |
Malassezia
|
Nitrososphaeria |
Polyomaviridae, Papillomaviridae, Circoviridae |
Acne53 |
Cutibacterium acnes (formerly Propionibacterium acnes) |
Firmicutes |
Atopic dermatitis54 |
Staphylococcus aureus
|
Bacteriodetes |
Proteobacteria |
Vagina55 |
Firmicutes (majorly Lactobacillus) |
Candida, uncultured Saccharomycetale |
Methanobacteria |
Bacteriophage |
Bacterial vaginosis56,57 |
Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., increased vaginal anaerobes decreased Lactobacillus spp. |
HIV infection58 |
Human immunodeficiency virus (HIV) |
HPV infection59 |
Human papillomavirus (HPV) |
Preterm birth60 |
Sneathia amnii, Lachnospiraceae, BVAB1, Saccharibacteria TM7-H1, decreased Lactobacillus crispatus |
3. The versatile toolbox of droplet microfluidics
Droplet microfluidics is a specialized branch of microfluidics that involves manipulating tiny, uniform droplets within immiscible carrier fluids.12,61,62 It emerged in the early 2000s and significantly advanced chemical and biological studies by enabling precise control of micro-reactions, facilitating high-throughput screening, and allowing single-cell analysis. The initial breakthrough came with the innovation of the T-junction device, which can produce monodispersed droplets through shear forces.63,64 This innovation enlightened subsequent droplet generation techniques, which enable a broad range of applications (Fig. 1).
 |
| Fig. 1 Overview of droplet microfluidic techniques. (A–C) Droplet generation using flow-focusing,65 step emulsification,66 and SlipChip.67 (D) Bifurcating droplet splitting.68 (E) Droplet fusion through a chamber of high electric field.69 (F) Droplet fusion within differential anchor array.70 (G) Off-chip droplet fusion by centrifugation in a 96-well microtiter plate.71 (H) Picoinjection.72 (I) Geometry-based trapping arrays.73 (J) Activated droplet sorting coupling different detection methods and external forces to select and collect target droplets.74 (K) Sorting and printing single-cell droplets based on fluorescence.75 (L) Integration of droplet microfluidics with flow cytometry and automated liquid handling for high-throughput screening.76 The schematics in B and L were created with https://www.BioRender.com. Panel C adapted from ref. 67 with permission, copyright 2020, Elsevier. | |
Droplet microfluidics offers several critical advantages, including precise control over reaction volumes, customizable mixing and reaction conditions,64 stochastic confinement of single cells or molecules,77 and minimal risk of cross-contamination.78 Its high throughput enables the rapid generation of droplets, making it ideal for large-scale biological analyses. Its flexibility in handling individual droplets enables a wide range of functional operations, such as splitting, mixing, and targeted sorting.62 These features make droplet microfluidics particularly valuable for single-cell analysis,79,80 microbial cultivation,81–84 nucleic acids amplification,85,86 and enzymatic reaction or screening.87,88 Given its versatility and scalability, understanding the various methods of droplet manipulation is essential for optimizing specific experimental outcomes. In the following sections, we will explore approaches for droplet generation and manipulation, highlighting their corresponding applications in biological research.
3.1 Droplet generation and incubation
Microfluidic droplet generation involves the continuous production of monodisperse droplets, typically ranging from picoliter to nanoliter volumes, by precisely shearing the dispersed phase with the continuous phase. Various methods have been developed for this process, including flow-focusing,65 step emulsification,66 capillary-based methods,89 and centrifugal-based techniques (Fig. 1A–C).90 Among these, flow-focusing and T-junction designs are the most widely used due to their simplicity, high throughput, and ability to generate droplets with sizes ranging from picoliters to microliters. Step emulsification, which uses parallel step structures, enables high-throughput droplet generation with minimal sensitivity to flow rate fluctuations. For instance, step emulsification chips can generate tens of thousands of droplets per second with sub-nanoliter volumes.66 Interfacial emulsification and OsciDrop leverage inertia forces using periodical vibrating capillaries or pipette tips to generate size-tunable droplets.91,92 Alternatively, SlipChip technology enables droplet generation by breaking a continuous stream into predefined volumes in microchambers following relative sliding of top and bottom plates, encapsulated by a continuous carrier fluid.67,93
Droplets can be incubated with well-controlled temperatures and atmospheres as uniform templates for spatially confined biochemical processes and advanced materials manufacturing.94 For example, picoliter to nanoliter droplets can undergo hours of thermal cycling without coalescence, enabling droplet digital PCR.92 On-chip incubation uses chambers integrated into the microfluidic device to provide precise environmental control,95 such as regulating oxygen levels to support the growth of anaerobes.96 In contrast, off-chip incubation is used for larger-scale or extended studies, where droplets are transferred to Eppendorf tubes or sealed environments to maintain stable conditions over time.97–99 Additionally, droplet microfluidics facilitates the creation of hydrogel microspheres from biocompatible polymers like agarose, alginate, or Matrigel, which encapsulate bacterial or mammalian cells for long-term growth with continuous media exchange.100 Droplets can also be tailored to study microscale events such as gene transfer, metabolic exchange, and biomolecular self-assembly.101
3.2 Droplet splitting, fusion, and picoinjection
Droplet splitting, fusion, and picoinjection are essential methods in droplet microfluidics that enable complex biological and chemical processes. Droplet splitting allows droplets to be divided symmetrically or asymmetrically into two or more smaller droplets (Fig. 1D). Symmetrical splitting is typically achieved by directing the droplet through bifurcating channels, where equal hydrodynamic resistance ensures the droplets divide evenly into two equal droplets.68
Droplet fusion involves merging two or more droplets to combine their contents, which is often needed in cascaded reactions where the mixing of reagents is necessary. In typical fusion devices, droplets of varying sizes are paired and compressed in a fusion chamber, where a high electric field facilitates their merging (Fig. 1E).69 While this method is highly efficient for contamination-sensitive and high-throughput applications, its success can be limited by imperfect matching droplet pairs. A more recent technique, the differential anchor array, selectively traps and immobilizes with different trapping strengths, allowing precise droplet pairing and fusion in a 2D array (Fig. 1F).70,102 Meanwhile, for on-demand droplet fusion off the chip, droplets can be dispensed in round-bottomed microtiter plates with carrier oil containing surfactant at the critical micelle concentration (CMC), followed by centrifugation, resulting in high fusion rates for single-cell assays (Fig. 1G).71
Picoinjection allows the precise introduction of picoliter volumes of reagents into individual droplets, also facilitated by an electric field, while maintaining droplet stability and integrity (Fig. 1H).72 A modified approach, stepinjection, utilizes a stepped injection channel design to balance injection success rates while minimizing contamination risks.103 Electrode-free picoinjection can be achieved by using dissolved electrolytes in the injection fluid as a conductor to apply the electric field.104 Alternatively, electricity-free picoinjection uses precise pressure control within the microfluidic channels to inject reagents without needing an electric field.105
3.3 Droplet trapping and array assembly
Droplets' trapping and array assembly are essential for prolonged observation, dynamic analysis, and rare event recovery. Automated detection systems can facilitate continuous and dynamic characterization by assigning positional information to each droplet fixed to a planar array. One commonly used approach involves geometry-based trapping designs, which utilize an array of microfabricated trap structures for high-throughput droplet capture (Fig. 1I).73 Hydrodynamic trapping chips guide droplets sequentially into chambers using fluid dynamics to enhance capture efficiency.106 Co-trapping of multiple droplets is another widely adopted strategy, essential for advanced applications requiring droplet interactions. For instance, the CARMEN (combinatorial arrayed reactions for multiplexed evaluation of nucleic acids)107 platform combines sample droplets with detection-mix droplets in a microwell array, enabling the random yet systematic pairing of diverse samples with amplification assays targeting various pathogens. This innovative approach achieves highly multiplexed target detection across numerous samples, significantly enhancing throughput and versatility in nucleic acid diagnostics.
Additionally, droplets can be assembled autonomously into planar monolayer droplet arrays (PMDA) within microfluidic chambers, creating large-scale arrays suitable for high-throughput imaging and analysis.108 Furthermore, in open reservoirs with flat bottoms, PMDAs can form through gravity-driven sedimentation and self-assembly, offering a streamlined method for organizing droplets on a large scale.91,92,109
3.4 Droplet sorting
Efficient sorting of droplets based on their properties is essential when processing large numbers of droplets. Sorting methods can be classified into passive and active techniques.74 Passive sorting leverages hydrodynamic properties to separate droplets based on factors like size or density.110,111 Passive sorting is cost-effective and scalable, ideal for high-throughput screening without external controls. This approach is particularly valuable in microbiome research, where bacterial growth within droplets can cause a size reduction,110 allowing the identification of droplets containing actively growing bacterial cells. In contrast, active sorting applies external forces such as dielectrophoresis,112 acoustic waves,113 magnetic forces,114 and pneumatic valves,115,116 to manipulate droplets on demand, overcoming flow resistance and directing selected droplets to collection channels. These platforms enable real-time sorting based on characteristics such as fluorescence intensity,117 absorbance,118,119 and real-time pattern recognition using high-speed imaging (Fig. 1J).120 Active droplet sorting has been widely used in applications such as antibody screening,121 drug-resistant strain screening,120 and bioactive molecules discovery.122
3.5 Integrated droplet microfluidics and downstream off-chip platforms
Integrating multiple droplet-based functional units reduces manual steps and enhances the efficiency of biochemical processes. Systems that combine droplet generation, incubation, and sorting within a single device have shown considerable progress in high-throughput applications. For example, one study reported a 30-fold increase in enzyme activity using a droplet microfluidic system that integrated single-cell lysis, enzymatic reactions, and fluorescence-activated droplet sorting (FADS).123 In another case, a microfluidic system combined droplet splitting and sorting modules for label-free screening using electrospray ionization mass spectrometry (ESI-MS). The droplets were split, with one portion analyzed by ESI-MS and the other sorted based on the results. This mass-activated droplet sorting (MADS) system achieved a throughput of 15
000 samples in 6 hours with an accuracy of 98% for in vitro-expressed transaminase.124
Additionally, integrating droplet microfluidics with other techniques can further expand its capabilities. For instance, combining droplet and digital microfluidics (DMF) enables automated sample preparation and high-throughput analyses.95,125 One integrated system performed nucleic acid extraction and reagent mixing in the DMF module, followed by droplet generation and collection in parallel chambers for multiplex digital loop-mediated isothermal amplification (dLAMP).125 This system demonstrated high accuracy (91.1%), speed, and a broad detection range for pathogen detection in urine samples. These innovations underscore the potential of integrated droplet microfluidic systems in advancing high-throughput biochemical analysis.
The potential of droplet microfluidics extends beyond miniaturized systems when integrated with established microtiter plate-based screening and liquid handling platforms, enabling a seamless transition from on-chip experiments to traditional laboratory workflows.126 For instance, the Cyto-Mine platform automates the encapsulation, screening, sorting, isolation, and dispensing of high-secreting clones into microtiter plate wells, achieving high precision and efficiency for monoclonality assurance.127 Similarly, a high-definition single-cell printing (HD-SCP) device was developed, featuring a miniaturized microfluidic sorter that selects single cells from a mixture based on fluorescence for printing with an accuracy of ∼10 μm and a speed of ∼100 Hz (Fig. 1K).75 This method has potential applications in printing bacterial cells encapsulated in droplets or hydrogel microspheres into microtiter plates, offering promising advancements for microbial studies and high-throughput screening. An alternative approach is transforming water-in-oil droplets into double emulsions, which can then be sorted using widely available flow cytometers (Fig. 1L).76 This method enables high-throughput screening of large droplet libraries, producing double emulsions at 6–12 kHz rates and sorting approximately 108 droplets in a day. Recently, a high-capacity approach developed with an optical-based microbubble technique has been applied to retrieve target droplets from a static droplet array (SDA).259 Unlike sorting at a fixed time point based on droplet content, this method utilizes a large-scale microchamber array to track real-time processes in each droplet simultaneously, allowing sorting based on reaction kinetics. This system showed nearly 100% success in releasing targeted droplets, proving its utility in high-throughput assays such as single-cell analysis and phenotype-based sorting.
4. Microbial single-cell sequencing
Microbial sequencing offers a culture-independent means of exploring microbial diversity and its functional roles in human health. However, traditional metagenomic methods always provide aggregate data from microbial populations, potentially masking key differences within subpopulations and overlooking rare microbes. Single-cell sequencing has emerged as a transformative approach for more detailed microbiome analysis and enabling the resolution of individual microbial cells (Fig. 2).
 |
| Fig. 2 Advances in droplet-based microbial single-cell sequencing. (A) Workflow of microbe-seq for microbial single-cell DNA sequencing.129 (B) Workflow of BacDrop for microbial scRNA-seq.130 (C) Integrating single-cell genome sequencing and metagenomics to improve genome completeness.131 (D) Analysis throughput (cell numbers per run) and genome coverage in representative publications for microbial single-cell genome sequencing. (E) Analysis throughput and transcriptome coverage (genes per cell) in representative publications for microbial scRNA-seq. | |
4.1 General methods in single-cell sequencing
Single cells contain genomic DNA in the femtogram range, making it challenging to extract enough material for direct sequencing. This problem can be solved by introducing whole-genome amplification (WGA) following single-cell lysis, which generates microgram quantities of DNA suitable for library preparation and sequencing. WGA techniques must carefully balance three key factors—amplification uniformity, genome coverage, and fidelity—to ensure reliable results, prompting the development of a range of innovative methods tailored to address these challenges. Early WGA techniques, such as degenerate oligonucleotide primer (DOP)-PCR132 combined with random primers, utilized polymerase chain reaction to amplify genomic DNA. Multiple displacement amplification (MDA)133 later emerged as a prominent method due to its use of Phi29 DNA polymerase with strand-displacement activity, enabling high-yield DNA amplification with improved coverage and accuracy. However, these exponential amplification-based methods can exacerbate amplification bias and amplification errors. To address these limitations, linear or semi-linear amplification approaches, like multiple annealing and looping-based amplification cycles (MALBAC)128 and linear amplification via transposon insertion (LIANTI),134 have improved amplification strategies to minimize bias and ensure uniform genome representation. Notably, LIANTI employs a transposon-based linear amplification principle, enabling precise detection of copy number variations and single nucleotide variations in human cells. However, its complex protocol has limited broader application on platforms like microfluidic systems. Recently, a derivative of MDA called primary template-directed amplification (PTA) has been introduced.135 By incorporating exonuclease-resistant terminators, PTA confines amplification primarily to the original template, achieving quasi-linear amplification. This approach not only maintains high amplification performance but also keeps simple experimental workflows. More detailed information about these aforementioned WGA techniques can be found in the review published by Wen et al.136
In single-cell RNA sequencing (scRNA-seq), the small amount of RNA in individual cells requires specialized techniques for capture and amplification. The process starts with single-cell lysis and RNA extraction, followed by reverse transcription into complementary DNA (cDNA). Techniques like Smart-seq,137,138 which use oligo-dT priming and template switching, generate full-length cDNA for amplification. Alternately, CEL-Seq139 utilizes oligo-dT primers integrated with T7 promoter elements and barcoding sequences, realizing linear amplification of cDNA through in vitro transcription and ensuring precise identification of transcripts originating from individual cells. Notably, the barcoding strategy140 allows sample pooling during library preparation, significantly improving throughput and reducing workload. These foundational techniques have laid the groundwork for advanced droplet-based single-cell sequencing platforms.
4.2 Advancements from traditional to droplet-based microbial single-cell sequencing
Early techniques for sequencing individual microbial cells relied on characteristics such as morphology, fluorescence labeling, or Raman spectroscopy, employing methods like flow cytometry,141 optical tweezers,142,143 and laser capture microdissection.144,145 These isolated cells were subjected to lysis, amplification, and library construction. Although these approaches had limited throughput, they successfully revealed genomic information from previously elusive microbial populations. More recently, high-throughput methods like PETRI-seq146 and microSPLiT147 have been developed, utilizing combinatorial barcoding to label permeabilized microbial cells in microtiter plates, enabling large-scale transcriptome sequencing. However, the multi-round barcoding process in these methods often results in significant data loss, making them less suitable for precious samples with limited cell numbers.
In contrast, droplet microfluidics has emerged as a transformative platform for single-cell multi-omics by transferring molecular reactions into individual droplets, offering high-throughput capabilities and efficient sample utilization. By enrichment of target microbial single cells through FADS, microbial genomes of rare species can be recovered with high quality.148 Encapsulating microbial single cells within gel beads enables in-bead cell lysis and whole-genome amplification in bulk systems, dramatically increasing operational throughput. Following fluorescence-activated droplet sorting (FACS) of gel beads, a large number of high-quality single-amplified genomes (SAGs) can be collected in plates for further research or storage.149 Furthermore, another similar study demonstrated that gel bead could achieve an average genome completeness significantly higher than that obtained using conventional microliter-scale amplification methods.150
Droplet microfluidics also enables the co-encapsulation of individual cells and barcoding beads within droplets, achieving in-droplet labeling to distinguish genetic material from different cells. Techniques such as Indrop151 and Drop-seq152 have leveraged this approach to achieve high-throughput single-cell transcriptomic sequencing in eukaryotic cells, laying the foundation for subsequent studies in microbial omics. Recently, several in-droplet cell labeling methods tailored for microbiological applications have been developed, employing diverse methodologies. To provide a clearer understanding of the available droplet-based platforms, we outline a general workflow for both genomic and transcriptomic analyses of prokaryotic cells, offering insights into their potential and applicability in microbial research. (Fig. 2A and B).
4.3 Droplet-based microbial single-cell genome sequencing
Microbial single-cell genomic sequencing has historically lagged behind eukaryotic applications. Droplet microfluidics has revolutionized whole-genome amplification (WGA) by introducing emulsion WGA (eWGA), which partitions single-cell genomic DNA into numerous picoliter droplets, ensuring uniform amplification gains across DNA fragments.153 This advancement enables high-fidelity analysis of low-input samples, such as single cells, for both eukaryotic and prokaryotic organisms.153,154 Recent innovations, such as SiC-seq and Microbe-seq, have further advanced the field by significantly increasing the throughput for microbial single-cell analysis.129,155 SiC-seq performs direct single-cell tagmentation and barcoding within droplets,155 while Microbe-seq integrates all processing steps into a droplet-based workflow, using MDA before genome labeling.129 This approach significantly enhances nucleic acid capture from single cells and improves genome coverage during sequencing (Fig. 2A). Microbe-seq has been used to reconstruct horizontal gene transfer networks in the gut microbiome and to identify host–phage interactions, such as the association between crAssphage and Bacteroides vulgatus.
Despite these advancements, SAGs often suffer from incomplete and uneven coverage due to artifacts introduced by MDA, prompting the development of co-assembly methods to generate more complete genome drafts such as ccSAG.156 This tool could effectively eliminate chimeric and co-assemble multiple and closely related SAG into a nearly complete genome with overall quality equivalent to those assembled from bulk DNA. Additionally, an approach integrating environmental metagenomic data has also been developed to overcome this bottleneck, which helps fill gaps within SAGs, significantly improving the completeness of genomes, such as those of SAR324 bacterioplankton and Thaumarchaeota.157 The bioinformatic tool SMAGLinker was designed to integrate SAG and metagenomic data to reconstruct single-cell genome-guided bins (sgBins), resulting in metagenome-guided SAGs (mgSAGs) with improved genome completeness compared to original SAGs (Fig. 2C).131
Recent single-cell technologies like DoTA-Seq focus on targeted gene profiling rather than whole-genome sequencing.158 This approach reduces experimental costs and time while enhancing data accuracy, making it ideal for detecting subtle expression changes in microbial communities and studying the role of low-abundance microbes in human disease. Additionally, SIP-seq has been developed to isolate and sequence HIV proviruses and their chromosomal integration sites at the single-cell level.159 This method employs FACS to select infected host cells, followed by two rounds of amplification—initially using MDA to amplify both viral and host DNA and then TaqMan PCR to detect HIV sequences. SIP-seq effectively eliminates interference from the host genome, allowing for precise identification of HIV provirus integration sites and offering insights into the clonal architecture and persistence of the HIV-1 reservoir.
4.4 Droplet-based microbial scRNA-seq
Transcriptomic sequencing is crucial in uncovering microbial diversity, heterogeneity, dynamic functions, and interactions in various environments.160 In microbial scRNA-seq, specific challenges arise compared to genome sequencing. Bacterial ribosomal RNA (rRNA) makes up over 80% of total RNA, while mRNA constitutes only a small fraction. Additionally, bacterial mRNA lacks the 5′ cap and PolyA tail found in eukaryotic mRNA, contributing to its instability and difficulty in capture.
Two key strategies have been developed to capture mRNA to address the challenges of prokaryotic scRNA-seq. The first approach uses random primers, which target a wide range of mRNA sequences. Technologies like scDual-Seq replaced poly(A)-based primers with random hexamers to capture eukaryotic RNA and synthesize the second strand using CEL-Seq2 barcoded primers.161 Recent technologies like BacDrop130 and smRandom-seq162 have further enhanced throughput, enabling the analysis of thousands of microbes per run. BacDrop depletes rRNA using a duplex-specific nuclease, followed by a two-step barcoding strategy (Fig. 2B).130 smRandom-seq uses the CRISPR-Cas9 system to selectively remove rRNA and applies short random primers for reverse transcription.162 The second approach employs gene-specific primers to improve the capture efficiency of specific mRNA sequences. For example, ProBac-seq was developed, utilizing large oligonucleotide libraries with sequences complementary to specific microbial genomes, pre-adenylated with PolyA tails, for targeted and efficient mRNA reverse transcription.163 This method has revealed previously unreported transcriptional heterogeneity in Escherichia coli (E. coli) and Bacillus subtilis (B. subtilis) and has provided insights into toxin expression regulation in Clostridium perfringens.
Dual scRNA-seq technologies have been developed to simultaneously monitor the expression profiles of eukaryotic cells and microbes at the single-cell level. For example, smRandom-seq has been used to study the interaction between Acinetobacter baumannii (A. baumannii) and macrophages, revealing a relationship between ferroptosis and A. baumannii infection.164 An unbiased dual-scRNA-seq method was applied to detect cells parasitized by Leishmania donovani, highlighting its ability to identify infected cells that were previously undetectable, such as hematopoietic stem cells in the bone marrow.165 Dual RNA-seq has also been employed to explore the tropism and receptor specificity of pandemic viruses.166 This approach provides valuable insights into microbial interactions within host cells and supports the development of targeted therapies for microbe-related diseases.
A comparative overview of major single-cell sequencing parameters for microbial genome sequencing (Fig. 2D) and scRNA-seq technologies (Fig. 2E). Emerging single-cell sequencing techniques are primarily based on droplet microfluidics platforms. While these platforms often yield lower per-cell data than traditional well-plate methods, they offer a significant advantage in throughput. Recent advances in droplet microfluidics have continuously improved genome coverage in DNA sequencing and increased the number of genes detected per cell in RNA sequencing. These advancements underscore the tremendous potential of this technology for a wide range of future applications. However, subtle variations in the microbial species and sequencing depths used across these methods imply that the parameters are more suited for general reference rather than direct comparison.
5. Droplet-based microbial isolation and cultivation
Microbial isolation aims to cultivate pure cultures to study microbial functions and their roles in human health. While culture-based methods have long been essential for classifying microbes by their physiological traits, modern culture-free approaches, such as metagenomics, have significantly expanded our understanding of microbial diversity by directly analyzing genetic material from natural environments. These methods have revealed numerous previously unknown microbes in the human gut,167 including archaea and viruses. However, despite their advantages, culture-independent methods face challenges like experimental and computational biases, emphasizing the continued need for pure cultures in functional studies and therapeutic applications. The culturomics approach has emerged as a powerful post-omics tool, leveraging advanced techniques to cultivate previously uncultivable microbial “dark matter”.168 Culturomics integrates high-throughput culturing methods with state-of-the-art sequencing and mass spectrometry to identify novel microbial species, significantly increasing the number of known bacteria from 2776 in 2018 to 3253 in 2020, with 301 new species attributed to this method.169 However, conventional colony-picking methods for isolating species still present drawbacks, such as fast-growing species dominating plates and inhibiting the growth of slower-growing microbes and the potential for mixed colonies, requiring time-consuming re-streaking.
Droplet microfluidics provides a highly efficient method for isolating complex microbial communities into individual cultures, offering scalability to cover diverse and low-abundance species while ensuring high purity and precision (Fig. 3). One example is the DropSpots device, which immobilizes thousands of picoliter-scale droplets, each containing a single microbial cell, for long-term cultivation and recovery (Fig. 3A).170 Another system, the large-scale droplet array in a microcage array, generates up to 1
000
000 droplets on a chip within seconds, facilitating microbial growth and enabling scalable downstream applications, such as screening esterase-expressing bacteria (Fig. 3B).171 The microfluidic streak plate (MSP) system arranges single-cell droplets in a spiral pattern for extended cultivation and recovery, significantly improving the growth of rare and slow-growing species (Fig. 3C).97,172 Droplet-based cultivation has also been employed to enhance the growth of strains typically found in low-abundance (<1%) in metagenomic analyses of human stool samples.120 Additionally, converting droplets into double emulsions allows them to be sorted via flow cytometry,173 facilitating nutrient privatization and enriching slow-growing species like Negativicutes and Methanobacteria from human stool samples.
 |
| Fig. 3 Strategies for isolating and cultivating difficult-to-culture microbes using droplet microfluidics. (A) DropSpots device for single-cell cultivation in a droplet array.170 (B) Microcage array chip for real-time microbial growth observation and droplet recovery.171 (C) Microfluidic streak plate for microbial cultivation.172 (D and E) Droplet co-encapsulation with essential symbiotic partners promotes microbial proliferation, as validated by imaging (D)174 and sequencing (E).96 (F) A 3D microfluidic chip integrates gradient dilution with droplet generation to adjust culture conditions.175 (G) Single-cell droplet dispensing systems for media optimization.176 (H) SlipChip for gene-targeted cultivation of microbes from the human gut.177 (I) epicPCR unveils episymbiotic relationships within microbial communities.178 Panel A adapted from ref. 170 with permission, copyright 2009, Royal Society of Chemistry. Panel B preprinted from ref. 171 with permission, copyright 2019, American Chemical Society. Panel C adapted from ref. 172 with permission, copyright 2016, American Society for Microbiology. Panel F reprinted from ref. 175 with permission, copyright 2015, American Chemical Society. | |
To investigate microbial interdependencies, such as cross-feeding and symbiotic relationships necessary for survival and proliferation, we can adjust the initial cell number based on the Poisson distribution of cells in droplets, facilitating the co-cultivation of symbiotic microbes.96,174 For example, this approach enabled the cultivation of previously uncharacterized gut commensals, such as a novel genus from the Neisseriaceae family, after successful proof-of-concept studies with synthetic auxotrophic E. coli strains (Fig. 3D). Another study used a high-throughput picoliter droplet system combined with microbial-derived necromass supplements to enrich candidate phyla radiation (CPR) bacteria from groundwater, highlighting its effectiveness for cultivating microbes with specialized parasitic or symbiotic relationships (Fig. 3E).179
To test bacterial growth under varying nutrient conditions, multilayer droplet microfluidic systems have been developed to generate concentration gradients and conduct high-throughput screenings (Fig. 3F).175 This method enhances the optimization of microbial culture environments, facilitating the evaluation of microbial responses to diverse conditions. Additionally, single-cell dispensers offer a precise alternative for testing cultivation conditions, with commercially available instruments enabling accurate droplet generation and cell isolation (Fig. 3G).176 Notably, a study demonstrated that combining bioorthogonal non-canonical amino acid tagging (BONCAT) with FACS effectively identifies translationally active microbes under various substrate and physicochemical conditions.180 This approach, paired with 16S rRNA sequencing for taxonomic identification, allows researchers to experimentally test genomic predictions. This method could also inform the development of targeted cultivation media for uncultured microbes, including archaea and bacteria, under conditions closely resembling their natural environments.
In addition to non-targeted isolation and cultivation methods, droplet microfluidics can be combined with techniques like Raman-activated cell sorting (RACS),181 optical tweezers,142 and fluorescence in situ hybridization (FISH)182 for precise capture and retrieval of target microbes based on phenotypes, genotypes, and surface proteins. For instance, a platform called Raman-activated gravity-driven single-cell encapsulation and sequencing (RAGE-Seq) was developed to identify antibiotic-resistant bacteria in urogenital tract infections.183 This platform uses single-cell Raman spectroscopy to phenotypically screen cells and then encapsulates the target cells in picoliter droplets for genomic sequencing. Raman spectroscopy detects molecular fingerprints unique to each species or cell based on the vibrational information of cellular components like DNA, proteins, and lipids, providing non-destructive, label-free sorting capabilities. Surface-enhanced Raman scattering (SERS) and stimulated Raman scattering (SRS) further enhance sensitivity and speed, enabling the detection of low-abundance molecules and rapid analysis for high-throughput applications.184,185
Another technique, the SlipChip device, employs two complementary plates with microchambers that split microcolonies into two daughter droplets for gene-targeted microbial isolation from a complex community (Fig. 3H). One half undergoes analysis via FISH or PCR, while the other is retained for cultivation, allowing successful isolation of elusive strains such as Oscillibacter from the human microbiome.177 Furthermore, the epicPCR (emulsion, paired isolation and concatenation PCR) technique,186,187 combined with droplet microfluidics,188 enables the isolation of episymbiotic bacteria like Candidatus Saccharibacteria (named TM7i) with its host (Leucobacter aridocollis) by linking phylogenetic markers to functional genes, facilitating the discovery of symbiotic relationships in microbial communities (Fig. 3I).178
Recent innovations in targeted microbial isolation also include a reverse genomics approach, where membrane protein-encoding genes are used to screen surface epitopes, and corresponding antibodies are created to capture previously uncultured Saccharibacteria along with their Actinobacteria hosts.189 Similarly, genomic information from the cell wall binding domain (CBD) of Streptococcus phages has been used to generate fluorescent CBDs to detect and enrich specific Streptococcus species from saliva.190 A digital microfluidics platform integrating optical tweezers and fluorescence-activated cell sorting was developed to capture Salmonella Typhimurium using magnetic beads coated with fluorescent-labeled antibiotics in droplets.191 This approach highlights the potential of combining reverse genomics with droplet microfluidics and other advanced techniques for isolating and cultivating previously unculturable microbes.
6. Applications of droplet microfluidics for assessing microbial activity and interactions
Droplet-based microfluidics provides a versatile platform for high-throughput and high-content microscale analysis, enabling a wide range of studies related to microbial activities and interactions. Recent studies have leveraged this technology to observe microbial growth, screen bioactive compounds, evaluate antimicrobial susceptibility, and investigate microbial cell–cell interactions.
6.1 Observing microbial growth and collective behavior
A microchemostat is a microscale version of a traditional chemostat designed to maintain a continuous and controlled environment for microbial growth. In a microchemostat, nutrients are continuously supplied, and waste products are removed, allowing microbes to grow under steady-state conditions. This setup is ideal for studying microbial behaviors over time, as it ensures consistent environmental conditions while enabling precise control over variables such as nutrient concentration, oxygen levels, and microbial density.
Integrated droplet microfluidic systems can manipulate each droplet as an individual microchemostat, providing a controlled environment for studying microbial growth dynamics at both individual and population levels.192 One study, for example, tracked the growth trajectory of hundreds of E. coli cells within droplets and demonstrated that the division times of individual cells were independent and uninherited, consistent with the Bellman–Harris model.193 Another study encapsulated the bacterial cell-division protein FtsZ into droplets, revealing self-organization behavior within confined environments, which provided new insights into the mechanisms of bacterial cell division.194 Also, microchemostats in droplet form have been used to study more complex microbial activities. For instance, a droplet-based study explored how droplet size and surface hydrophobicity influenced bacterial plasmid transfer rates in microscopic surface wetness (MSW) environments (Fig. 4A).101 The findings indicated that larger droplets with lower liquid–solid interface ratios had higher cell densities and increased plasmid transfer rates, while surface hydrophobicity further enhanced transfer rates by reducing droplet spreading and promoting higher cell densities.
 |
| Fig. 4 Droplet microfluidics for assessing microbial activity and interactions. (A) Droplets with smaller sizes promote plasmid transfer in the microbial population.101 (B) Single-cell quorum sensing activated in droplets.195 (C) Microbial biofilm formation in sessile droplets for long-term studies.196 (D) Multiplex ddPCR for amplification-based microbial identification.197 (E) Amplification-free microbial identification using DNAzyme-based sensors.198 (F) Microbial antibiotic susceptibility testing conducted via the DropFAST platform.199 (G) AI-driven analysis platform for determining optimal antibiotic combinations based on high-throughput imaging and droplet content analysis.200 (H) MINI-Drop method co-encapsulates multiple microbial species to investigate their interactions.201 (I) The kChip for high-throughput and random fusion of droplets containing different microbial species. It allows analysis of growth dynamics and screening of synthetic microbial communities.108 Panel B reprinted from ref. 195 with permission, copyright 2009, WILEY VCH. Panel C reprinted from ref. 196 with permission, copyright 2009, WILEY VCH. Panel G reprinted from ref. 200 with permission, copyright 2023, Royal Society of Chemistry. | |
Droplet-based microchemostats are useful for studying isolated microbial growth and investigating collective microbial behaviors, such as quorum sensing and biofilm formation. For example, individual Staphylococcus aureus (S. aureus) cell in droplets was found to secrete autoinducer that initiated quorum sensing, as the confined droplet environment concentrated quorum-sensing factors, thereby activating regulatory genes (Fig. 4B).195 A dynamic sessile-droplet system was developed to cultivate and analyze P. aeruginosa biofilms on hydrophilic micropatterns under shear force stimulation, providing a robust platform for studying biofilm development and testing antimicrobial susceptibility (Fig. 4C).196
6.2 Screening microbial bioactive molecules
Microbial genomes are incredibly diverse, offering various functional capabilities that allow microbes to catalyze numerous biochemical processes.202,203 This diversity makes microbes valuable for discovering novel enzymes, metabolites, and bioactive compounds. Droplet-based microfluidics, particularly when combined with FADS or emulsified into double emulsions for use with traditional FACS, has emerged as a powerful tool for high-throughput screening of bioactive molecules.
Droplet microfluidics facilitates the detection of secreted products of microbial cells within picoliter droplets, preserving the critical genotype–phenotype linkage essential for screening in metabolic engineering.88,122 For instance, a transcription-based E. coli biosensor, responsive to p-coumaric acid via the B. subtilis transcriptional repressor PadR, was used to sort yeast cells encapsulated in droplets based on their extracellular p-coumaric acid production, indicated by a PadR-regulated fluorescent signal.204 RNA aptamer technology has been used to reflect target molecule concentrations, such as tyrosine, into fluorescence signals, enabling ultra-high-throughput screening of millions of yeast mutants. This method's adaptability for various target molecules through aptamer sequence modification enhances its versatility for bioactive molecule screening.205 FADS has also been applied to detect antibiotic-producing microbes within microbial communities, identifying various antimicrobial compounds like surfactants and cytotoxic lipopeptides.206 Additionally, a solid-state fermentation medium with 2% colloidal chitin was employed to fortify droplet structural integrity, preventing hyphae penetration and supporting long-term fungal cultivation, facilitating FADS-based screening of clones with high cell-wall-degrading enzymatic activity.207 Furthermore, water–oil–water double emulsions were used in FACS to screen riboflavin-secreting mutant cells, achieving riboflavin production rates 56 times higher than the parent strain.208
Integrating microfluidic systems with advanced mass spectrometric techniques, such as electrospray ionization (ESI) and matrix-assisted laser desorption ionization (MALDI), has revolutionized the analysis of microbial metabolites.209,210 These methods offer high sensitivity and resolution for detecting metabolites at the single-cell level. For example, one study combined droplet microfluidics with ESI-MS to detect secondary metabolites like streptomycin from actinomycetales strains,211,212 providing a robust tool for discovering novel antibiotics. Further innovations, such as using glass chips for droplet generation and fluorinated capillaries to eliminate surfactant interference in ESI-MS detection,212,213 have facilitated biocatalysis detection at single-cell resolution. These droplet-MS detection techniques eliminate the need for labeling and streamline the discovery of functional microbial metabolites.
6.3 Droplet-based pathogen diagnostics and antimicrobial susceptibility testing
Antimicrobial resistance (AMR) poses a global health threat, with estimates suggesting it could result in 10 million deaths annually by 2050.214 Delays in cultivation, pathogen identification, and antimicrobial susceptibility testing (AST) contribute to the overuse and misuse of antibiotics, exacerbating the AMR crisis. Droplet microfluidics offers a promising solution by facilitating faster pathogen cultivation, identification, and AST, thereby enabling precise antibiotic therapy and reducing the risks of treatment delays and inappropriate antibiotic use.
Droplet-based pathogen identification methods offer rapid detection, high sensitivity, and efficient processing of complex clinical samples. By compartmentalizing samples into thousands of droplets, droplet digital PCR (ddPCR) enables highly sensitive and multiplexed detection of methicillin-resistant Staphylococcus aureus (MRSA), even at low copy numbers, which is crucial for diagnosing early-stage infections (Fig. 4D). Compared to qPCR, ddPCR demonstrated significantly superior sensitivity (100% vs. 38.89%) and comparable or higher specificity (100% vs. 97.67%) in detecting MRSA.197 Furthermore, in another comparative study, ddPCR exhibited greater precision and robustness than qPCR in quantifying Cryptosporidium oocysts from fecal samples, particularly under challenging conditions such as low DNA concentrations and the presence of PCR inhibitors.215 Additionally, ddPCR played a critical role during the COVID-19 pandemic, assisting in the detection of low-copy-number viruses, viral load quantification, and monitoring viral concentrations in the environment.216
Amplification-free approaches, such as DNAzyme-based sensors, have also been integrated into droplets for rapid and specific bacterial detection directly from clinical blood samples (Fig. 4E).198 Similarly, a droplet microfluidics system has been developed for the rapid diagnosis of urinary tract infections (UTIs) by using fluorogenic peptide nucleic acid (PNA) probes targeting 16S rRNA, allowing for amplification-free detection.217 In antimicrobial susceptibility testing, ddPCR can assess susceptibility after 15 minutes of antibiotic exposure by measuring DNA replication through single-molecule counting, offering a rapid, phenotype-based approach to UTI testing.218 Similarly, droplet digital loop-mediated isothermal amplification (ddLAMP) has been applied to determine E. coli susceptibility from clinical urine samples in less than 30 minutes, achieving accuracy comparable to standard methods.219
Droplet-based microfluidics provides additional benefits in AST by enabling faster detection, enhanced sensitivity, and high-throughput screening of antibiotic combinations.220 For example, a rapid single-cell biosensing platform, dropFAST, was developed to encapsulate individual bacterial cells with a fluorescent growth indicator and antibiotics into picoliter droplets (Fig. 4F). After a short on-chip incubation, in-line fluorescence detection yields AST results within 1 hour.199 This single-cell analysis enhances the accuracy of minimum inhibitory concentration (MIC) determinations by eliminating population-level interactions and nutrient competition, providing insights into antibiotic resistance mechanisms in clonally identical bacterial populations. A cascade filtration-droplet digital chromogenic assay system was also developed to identify carbapenem-resistant strains in urine samples without traditional isolation, achieving rapid identification by chromogenic reaction within 3.5 hours.221 Furthermore, machine learning-based morphological recognition has been integrated with droplet microfluidics to automate the classification of bacterial growth within nanoliter droplets,222,223 enabling the determination of colistin susceptibility profiles within 3 hours in a label-free manner and demonstrated similar accuracy to the traditional broth dilution method. Additionally, droplet microfluidics facilitates the assessment of antibiotic combinations, helping to optimize therapy, minimize side effects, and slow the evolution of multi-antibiotic-resistant bacterial strains (Fig. 4G).200
6.4 Investigating microbial interactions
In natural environments, microbes rarely exist in isolation, instead engaging in complex interactions with other species. This is equally true within the human body, where microbial interactions can significantly influence health and disease. These interactions generally fall into three categories: positive, negative, or neutral, based on phenotypic differences such as growth response or metabolic activity. However, despite advancements in bioinformatics tools like CoNet,224 MetagenoNets,225 and NetCoMi,226 there remains a notable gap between in silico predictions and experimental outcomes in microbial interactions.
Droplet-based microfluidics has appeared as an effective tool for studying microbial interactions, offering greater precision and efficiency than traditional culture methods.227 For instance, programmable droplet microfluidic devices have been designed to enable sequential merging of picoliter-volume droplets containing bacterial cells, allowing researchers to model complex microbial communities and study their stability and coexistence over time.228 These systems help study symbiotic relationships, such as cross-feeding auxotrophs. One example involved co-culturing an RFP-labeled E. coli methionine auxotroph with a GFP-labeled B. subtilis tryptophan auxotroph, enabling the detailed study of positive microbial interactions (Fig. 4H).201 Negative interactions have also been explored using droplet-based methods, such as a GFP-labeled E. coli strain producing a quorum-sensing molecule that inhibited another RFP-labeled E. coli strain. Further innovations include platforms like kChip, which can rapidly screen ∼100
000 synthetic microbial consortia for growth promoters or inhibitors, providing valuable insights into species interactions, even under varying carbon sources (Fig. 4I).108
Microfluidic devices are not limited to studying interactions within microbial species but are also applied to interkingdom interactions, such as those between bacteria and fungi or phages and bacteria. For example, time-lapse fluorescence microscopy has been combined with droplet microfluidics to observe the growth and lysis dynamics of E. coli when challenged with phages.229 In another study, a micrometabolomics device was used to demonstrate that Aspergillus fumigatus growth was inhibited in the presence of P. aeruginosa, while Aspergillus flavus exhibited enhanced chlamydospore formation when co-cultured with Ralstonia solanacearum.230 These applications highlight the versatility of droplet microfluidics in uncovering a broad range of microbial interactions.
7. Application of droplet-microfluidics for investigating the role of microbes on human health
Research on microbe–host interactions has gained significant attention for its role in understanding human health. These interactions are crucial for the colonization and evolution of microbial communities and for regulating host physiology and immune responses. Traditionally, animal models, such as germ-free and pseudo germ-free mice, have been instrumental in studying these interactions. For example, germ-free mice have been used to demonstrate that certain microbial species can improve intestinal barrier function, modulate immune responses, or trigger diseases like intestinal inflammation and gastric cancer.231,232
Building on these insights, microbial therapies to deliver or cut specific microbes for disease treatment have garnered increasing interest. Droplet microfluidics, with its advantages in precise size control, high-throughput production, and efficient multilayer design, has been applied to encapsulate and target microbes and their byproducts. For instance, Bacteroides fragilis, enriched in breast cancer tissues, was utilized to create mineralized bacterial outer membrane vesicles using a microfluidic platform, enhancing antitumor immune responses when delivered to tumor tissues.233 Similarly, microfluidic systems have been developed to co-encapsulate probiotics and postbiotics in double hydrogel droplets, targeting delivery to the colon for treating colitis by reducing inflammation and promoting intestinal barrier repair.234 Furthermore, advanced colloidosome platforms with multiple protective layers were designed to enhance probiotic survival during gastrointestinal transit, allowing for targeted release in the colon with significant therapeutic effects on colitis.235,236
In addition to animal models, in vitro models play a crucial role in studying microbe–host interactions. Traditional two-dimensional (2D) cell cultures often fail to accurately mimic physiological conditions. Advances in tissue engineering have led to the development of three-dimensional (3D) culture systems, such as spheroids and organoids, which better replicate tissue microenvironments. Droplet-based microfluidics has created controlled environments for 3D cell growth and interactions through scaffold-free or scaffold-based methods. For example, scaffold-free droplet microfluidics has been used to generate multicellular spheroids for drug testing,237 while scaffold-based methods incorporate extracellular matrix materials to study cell–matrix interactions and cellular differentiation.238
Despite these advances, droplet microfluidic devices have been less commonly applied to microbe–host interaction studies than human organ-on-chips systems.239 This is primarily due to the latter's ability to more accurately replicate the complex physiological environments of human organs, including tissue architectures, fluid dynamics, and multi-organ interactions, such as the gut–liver axis.240–244 Organ-on-a-chip systems also offer a continuous supply of nutrients and oxygen, which is crucial for long-term culture and monitoring of interactions. While droplet microfluidics has not yet reached this level of control, some analogous strategies have been developed. For instance, droplet pairing and fusion allow fresh medium to be introduced, mimicking fed-batch culture modes to some extent. Additionally, replacing the oil phase with culture media facilitates constant nutrient refreshment for immobilized cells within hydrogels, akin to continuous culture modes seen in bioreactors.100,245 Such advancements highlight the potential for droplet microfluidics to evolve toward more robust and versatile platforms for modeling microbe–host interactions in the future.
8. Conclusion and outlook
Advances in multidisciplinary fields, particularly meta-omics technologies, have transformed human microbiome research from focusing solely on microbial composition to uncovering functional relationships between the microbiome and host health. Emerging evidence highlights that microbiota-derived bioactive compounds, including antigens, enzymes, small molecules, and extracellular vesicles, play essential roles in regulating both local and systemic physiological pathways. Despite these advances, the precise mechanisms by which individual microbial taxa influence host gene expressions and pathways remain largely unknown.
Droplet microfluidics has emerged as a transformative tool in microbiome research, enabling high-throughput isolation, cultivation, and analysis of individual microbial cells within picoliter to nanoliter droplets. This approach benefits both culture-dependent and culture-independent studies by enabling precise manipulation of microenvironments or reactions. Despite these advancements, several challenges remain. Variability in single-cell sequencing across different microbial species highlights the need for standardized preprocessing methods that can be applied to a broader range of microbes. Additionally, sequencing coverage and the amount of usable data are still limited, calling for improvements in molecular techniques, such as enhanced nucleic acid capture and amplification. Single-cell proteomics and metabolomics in microbes are still underdeveloped, and the integration of droplet-based multi-omics technologies to explore causal relationships between microbes and host phenotypes is in its preliminary stages. Further innovations in droplet-based spatial omics are also needed to better elucidate microbial spatial organization and interactions with their environments.
Despite its potential, droplet microfluidics also faces several limitations compared to traditional methods. Microbial cultivation within droplets can sometimes fail to replicate the complex interactions present in bulk cultures, potentially affecting the growth of cross-feeding species. Additionally, operational complexity and the lack of standardized interfaces hinder its accessibility for researchers with limited expertise in microfluidics. Droplet stability issues further complicate multistep, intricate reactions. Furthermore, the limited integration of droplet microfluidics with high-throughput automated platforms restricts its full potential for broader applications. These challenges underscore the need for automation and standardization of microfluidic device production and operation to enable scalable applications.
Recent advances in artificial intelligence (AI) offer promising solutions to these challenges. AI has demonstrated its ability to enhance device design,246,247 sorting,248 and functional analysis in droplet microfluidics.249,250 AI-driven tools, including machine learning, are being used to optimize microbial media formulations,251–253 perform image-based microbial identification,254 and analyze multi-omics data.255 Looking ahead, the convergence of large-scale experimental data generated through droplet microfluidics and AI-driven analytics holds the potential to revolutionize human microbiome research. By enabling high-resolution, large-scale investigations, these technologies can uncover novel therapeutic strategies that harness the microbiome's potential for improving human health.256–258 This approach paves the way for transitioning from understanding microbial diversity to unlocking microbial functionality, driving advancements in precision medicine and the development of innovative treatments for microbe-associated diseases.
Data availability
No new data were generated or analyzed in this study. Data sharing is not applicable to this article as it is a review of existing literature.
Author contributions
Y. X. and Z. W. contributed equally to drafting and visualization of the original manuscript. C. L. contributed to visualization. S. T. was involved in investigation and editing. W. D. provided supervision, contributed to conceptualization, editing, and visualization.
Conflicts of interest
The authors declared no conflict of interest.
Note added after first publication
This review has been updated following first publication on 8th January 2025 to include ref. 259 at the request of the authors.
Acknowledgements
This work was supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB0830000), the National Natural Science Foundation of China (92251302), and the National Key Research and Development Program of China (2021YFC2301000, 2022YFA1304100, 2021YFA0717000).
References
- L. V. Hooper and J. I. Gordon, Science, 2001, 292, 1115–1118 CrossRef CAS PubMed.
- J. Qin, R. Li, J. Raes, M. Arumugam, K. S. Burgdorf, C. Manichanh, T. Nielsen, N. Pons, F. Levenez, T. Yamada, D. R. Mende, J. Li, J. Xu, S. Li, D. Li, J. Cao, B. Wang, H. Liang, H. Zheng, Y. Xie, J. Tap, P. Lepage, M. Bertalan, J.-M. Batto, T. Hansen, D. Le Paslier, A. Linneberg, H. B. Nielsen, E. Pelletier, P. Renault, T. Sicheritz-Ponten, K. Turner, H. Zhu, C. Yu, S. Li, M. Jian, Y. Zhou, Y. Li, X. Zhang, S. Li, N. Qin, H. Yang, J. Wang, S. Brunak, J. Doré, F. Guarner, K. Kristiansen, O. Pedersen, J. Parkhill, J. Weissenbach, MetaHIT Consortium, P. Bork, S. D. Ehrlich and J. Wang, Nature, 2010, 464, 59–65 CrossRef CAS PubMed.
- P. J. Turnbaugh, R. E. Ley, M. Hamady, C. M. Fraser-Liggett, R. Knight and J. I. Gordon, Nature, 2007, 449, 804–810 CrossRef CAS PubMed.
- The Integrative HMP (iHMP) Research Network Consortium, Nature, 2019, 569, 641–648 CrossRef PubMed.
- D. Vandeputte, L. De Commer, R. Y. Tito, G. Kathagen, J. Sabino, S. Vermeire, K. Faust and J. Raes, Nat. Commun., 2021, 12, 6740 CrossRef CAS PubMed.
- D. Zheng, T. Liwinski and E. Elinav, Cell Res., 2020, 30, 492–506 CrossRef PubMed.
- Y. Zhen, Y. Wang, F. He, Y. Chen, L. Hu, L. Ge, Y. Wang, W. Wei, A. Rahmat, J. J. Loor and M. Wang, J. Pineal Res., 2023, 75, e12892 CrossRef CAS PubMed.
- T. Wilmanski, S. A. Kornilov, C. Diener, M. P. Conomos, J. C. Lovejoy, P. Sebastiani, E. S. Orwoll, L. Hood, N. D. Price, N. Rappaport, A. T. Magis and S. M. Gibbons, Med, 2022, 3, 388–405.E6 CrossRef CAS PubMed.
- B. Youenou, S. Favre-Bonté, J. Bodilis, E. Brothier, A. Dubost, D. Muller and S. Nazaret, Genome Biol. Evol., 2015, 7, 2484–2505 CrossRef CAS PubMed.
- H. P. Browne, S. C. Forster, B. O. Anonye, N. Kumar, B. A. Neville, M. D. Stares, D. Goulding and T. D. Lawley, Nature, 2016, 533, 543–546 CrossRef CAS PubMed.
- E. K. Sackmann, A. L. Fulton and D. J. Beebe, Nature, 2014, 507, 181–189 CrossRef CAS PubMed.
- L. Nan, H. Zhang, D. A. Weitz and H. C. Shum, Lab Chip, 2024, 24, 1135–1153 RSC.
- The Human Microbiome Project Consortium, Nature, 2012, 486, 207–214 CrossRef PubMed.
- J. A. Gilbert, M. J. Blaser, J. G. Caporaso, J. K. Jansson, S. V. Lynch and R. Knight, Nat. Med., 2018, 24, 392–400 CrossRef CAS PubMed.
- D. Bogaert, G. J. Van Beveren, E. M. De Koff, P. L. Parga, C. E. B. Lopez, L. Koppensteiner, M. Clerc, R. Hasrat, K. Arp, M. L. J. N. Chu, P. C. M. De Groot, E. A. M. Sanders, M. A. Van Houten and W. A. A. De Steenhuijsen Piters, Cell Host Microbe, 2023, 31, 447–460.e6 CrossRef CAS PubMed.
- R. E. Moore and S. D. Townsend, Open Biol., 2019, 9, 190128 CrossRef CAS PubMed.
- A. T. Neu, E. E. Allen and K. Roy, Proc. Natl. Acad. Sci. U. S. A., 2021, 118, e2104429118 CrossRef CAS PubMed.
- G. Wu, T. Xu, N. Zhao, Y. Y. Lam, X. Ding, D. Wei, J. Fan, Y. Shi, X. Li, M. Li, S. Ji, X. Wang, H. Fu, F. Zhang, Y. Shi, C. Zhang, Y. Peng and L. Zhao, Cell, 2024, 187, 6550–6565 CrossRef CAS PubMed.
- N.-N. Liu, N. Jiao, J.-C. Tan, Z. Wang, D. Wu, A.-J. Wang, J. Chen, L. Tao, C. Zhou, W. Fang, I. H. Cheong, W. Pan, W. Liao, Z. Kozlakidis, C. Heeschen, G. G. Moore, L. Zhu, X. Chen, G. Zhang, R. Zhu and H. Wang, Nat. Microbiol., 2022, 7, 238–250 CrossRef CAS PubMed.
- Q. Su, O. W. H. Wong, W. Lu, Y. Wan, L. Zhang, W. Xu, M. K. T. Li, C. Liu, C. P. Cheung, J. Y. L. Ching, P. K. Cheong, T. F. Leung, S. Chan, P. Leung, F. K. L. Chan and S. C. Ng, Nat. Microbiol., 2024, 9, 2344–2355 CrossRef CAS PubMed.
- M. Arumugam, J. Raes, E. Pelletier, D. Le Paslier, T. Yamada, D. R. Mende, G. R. Fernandes, J. Tap, T. Bruls, J.-M. Batto, M. Bertalan, N. Borruel, F. Casellas, L. Fernandez, L. Gautier, T. Hansen, M. Hattori, T. Hayashi, M. Kleerebezem, K. Kurokawa, M. Leclerc, F. Levenez, C. Manichanh, H. B. Nielsen, T. Nielsen, N. Pons, J. Poulain, J. Qin, T. Sicheritz-Ponten, S. Tims, D. Torrents, E. Ugarte, E. G. Zoetendal, J. Wang, F. Guarner, O. Pedersen, W. M. de Vos, S. Brunak, J. Doré, MetaHIT Consortium (additional members), J. Weissenbach, S. D. Ehrlich and P. Bork, Nature, 2011, 473, 174–180 CrossRef CAS PubMed.
- P. Lemanceau, M. Blouin, D. Muller and Y. Moënne-Loccoz, Trends Plant Sci., 2017, 22, 583–595 CrossRef CAS PubMed.
- A. Shade and J. Handelsman, Environ. Microbiol., 2012, 14, 4–12 CrossRef CAS PubMed.
- K. Li, M. Bihan and B. A. Methé, PLoS One, 2013, 8, e63139 CrossRef CAS PubMed.
- L. Cui, A. Morris and E. Ghedin, Genome Med., 2013, 5, 63 CrossRef PubMed.
- P. Belvoncikova, P. Splichalova, P. Videnska and R. Gardlik, J. Fungi, 2022, 8, 1046 CrossRef PubMed.
- P. Geesink and T. J. G. Ettema, Nat. Microbiol., 2021, 7, 10–11 CrossRef PubMed.
- T. Kuehnast, C. Kumpitsch, R. Mohammadzadeh, T. Weichhart, C. Moissl-Eichinger and H. Heine, FEBS J., 2025, 292 DOI:10.1111/febs.17123 , in press.
- G. Liang and F. D. Bushman, Nat. Rev. Microbiol., 2021, 19, 514–527 CrossRef CAS PubMed.
- A. K. Nash, T. A. Auchtung, M. C. Wong, D. P. Smith, J. R. Gesell, M. C. Ross, C. J. Stewart, G. A. Metcalf, D. M. Muzny, R. A. Gibbs, N. J. Ajami and J. F. Petrosino, Microbiome, 2017, 5, 153 CrossRef PubMed.
- W. Panpetch, P. Hiengrach, S. Nilgate, S. Tumwasorn, N. Somboonna, A. Wilantho, P. Chatthanathon, P. Prueksapanich and A. Leelahavanichkul, Gut Microbes, 2020, 11, 465–480 CrossRef PubMed.
- R. S. Dalal and J. R. Allegretti, Curr. Opin. Gastroenterol., 2021, 37, 336–343 CrossRef CAS PubMed.
- S. Federici, S. Kredo-Russo, R. Valdés-Mas, D. Kviatcovsky, E. Weinstock, Y. Matiuhin, Y. Silberberg, K. Atarashi, M. Furuichi, A. Oka, B. Liu, M. Fibelman, I. N. Weiner, E. Khabra, N. Cullin, N. Ben-Yishai, D. Inbar, H. Ben-David, J. Nicenboim, N. Kowalsman, W. Lieb, E. Kario, T. Cohen, Y. F. Geffen, L. Zelcbuch, A. Cohen, U. Rappo, I. Gahali-Sass, M. Golembo, V. Lev, M. Dori-Bachash, H. Shapiro, C. Moresi, A. Cuevas-Sierra, G. Mohapatra, L. Kern, D. Zheng, S. P. Nobs, J. Suez, N. Stettner, A. Harmelin, N. Zak, S. Puttagunta, M. Bassan, K. Honda, H. Sokol, C. Bang, A. Franke, C. Schramm, N. Maharshak, R. B. Sartor, R. Sorek and E. Elinav, Cell, 2022, 185, 2879–2898.e24 CrossRef CAS PubMed.
- M. Jayachandran and S. Qu, Rev. Endocr. Metab. Disord., 2023, 24, 1189–1204 CrossRef PubMed.
- T. Yu, F. Guo, Y. Yu, T. Sun, D. Ma, J. Han, Y. Qian, I. Kryczek, D. Sun, N. Nagarsheth, Y. Chen, H. Chen, J. Hong, W. Zou and J.-Y. Fang, Cell, 2017, 170, 548–563.e16 CrossRef CAS PubMed.
- R. Li, J. Li and X. Zhou, Signal Transduction Targeted Ther., 2024, 9, 19 CrossRef PubMed.
- K. Koskinen, M. R. Pausan, A. K. Perras, M. Beck, C. Bang, M. Mora, A. Schilhabel, R. Schmitz and C. Moissl-Eichinger, mBio, 2017, 8, e00824-17 CrossRef PubMed.
- M. F. Moffatt and W. O. Cookson, Clin. Med., 2017, 17, 525–529 CrossRef PubMed.
- A. A. Abbas, L. J. Taylor, M. I. Dothard, J. S. Leiby, A. S. Fitzgerald, L. A. Khatib, R. G. Collman and F. D. Bushman, Cell Host Microbe, 2019, 25, 719–729.e4 CrossRef CAS PubMed.
- Z. Yan, B. Chen, Y. Yang, X. Yi, M. Wei, G. Ecklu-Mensah, M. M. Buschmann, H. Liu, J. Gao, W. Liang, X. Liu, J. Yang, W. Ma, Z. Liang, F. Wang, D. Chen, L. Wang, W. Shi, M. R. Stampfli, P. Li, S. Gong, X. Chen, W. Shu, E. M. El-Omar, J. A. Gilbert, M. J. Blaser, H. Zhou, R. Chen and Z. Wang, Nat. Microbiol., 2022, 7, 1361–1375 CrossRef CAS PubMed.
- S. Knippenberg, B. Ueberberg, R. Maus, J. Bohling, N. Ding, M. Tort Tarres, H.-G. Hoymann, D. Jonigk, N. Izykowski, J. C. Paton, A. D. Ogunniyi, S. Lindig, M. Bauer, T. Welte, W. Seeger, A. Guenther, T. H. Sisson, J. Gauldie, M. Kolb and U. A. Maus, Thorax, 2015, 70, 636–646 CrossRef PubMed.
- P. M. George, A. U. Wells and R. G. Jenkins, Lancet Respir. Med., 2020, 8, 807–815 CrossRef CAS PubMed.
- M. Richardson, P. Bowyer and R. Sabino, Med. Mycol., 2019, 57, S145–S154 CrossRef PubMed.
- M. A. Behr, E. Kaufmann, J. Duffin, P. H. Edelstein and L. Ramakrishnan, Am. J. Respir. Crit. Care Med., 2021, 204, 142–148 CrossRef PubMed.
- M. A. Ghannoum, R. J. Jurevic, P. K. Mukherjee, F. Cui, M. Sikaroodi, A. Naqvi and P. M. Gillevet, PLoS Pathog., 2010, 6, e1000713 CrossRef PubMed.
- D. Verma, P. K. Garg and A. K. Dubey, Arch. Microbiol., 2018, 200, 525–540 CrossRef CAS PubMed.
- Y.-Y. Li, Q. Cai, B.-S. Li, S.-W. Qiao, J.-Y. Jiang, D. Wang, X.-C. Du and W.-Y. Meng, Inflammation, 2021, 44, 846–858 CrossRef CAS PubMed.
- Y. Lu, Y. Lin, M. Li and J. He, Front. Cell. Infect. Microbiol., 2023, 13, 1151532 CrossRef CAS PubMed.
- J.-L. Zhang, J. Yun, L. Yue, W. Du and Y.-H. Liang, Front. Cell. Infect. Microbiol., 2022, 12, 980157 CrossRef CAS PubMed.
- M. M. Elashiry, B. E. Bergeron and F. R. Tay, Microb. Pathog., 2023, 183, 106337 CrossRef CAS PubMed.
- A. L. Byrd, Y. Belkaid and J. A. Segre, Nat. Rev. Microbiol., 2018, 16, 143–155 CrossRef CAS PubMed.
- A. J. Probst, A. K. Auerbach and C. Moissl-Eichinger, PLoS One, 2013, 8, e65388 CrossRef CAS PubMed.
- T. Yu, X. Xu, Y. Liu, X. Wang, S. Wu, Z. Qiu, X. Liu, X. Pan, C. Gu, S. Wang, L. Dong, W. Li and X. Yao, Cell Host Microbe, 2024, 32, 1129–1146.e8 CrossRef CAS PubMed.
- M. C. Moran, M. P. Cahill, M. G. Brewer, T. Yoshida, S. Knowlden, N. Perez-Nazario, P. M. Schlievert and L. A. Beck, mSphere, 2019, 4, e00616–e00619 CrossRef CAS PubMed.
- J. Ravel, P. Gajer, Z. Abdo, G. M. Schneider, S. S. K. Koenig, S. L. McCulle, S. Karlebach, R. Gorle, J. Russell, C. O. Tacket, R. M. Brotman, C. C. Davis, K. Ault, L. Peralta and L. J. Forney, Proc. Natl. Acad. Sci. U. S. A., 2011, 108, 4680–4687 CrossRef CAS PubMed.
- L. Huang, R. Guo, S. Li, X. Wu, Y. Zhang, S. Guo, Y. Lv, Z. Xiao, J. Kang, J. Meng, P. Zhou, J. Ma, W. You, Y. Zhang, H. Yu, J. Zhao, G. Huang, Z. Duan, Q. Yan and W. Sun, Nat. Microbiol., 2024, 9, 2185–2200 CrossRef CAS PubMed.
- C. A. Muzny, C. M. Taylor, W. E. Swords, A. Tamhane, D. Chattopadhyay, N. Cerca and J. R. Schwebke, J. Infect. Dis., 2019, 220, 1399–1405 CrossRef CAS PubMed.
- E. M. Cherenack, C. A. Broedlow and N. R. Klatt, Curr. Opin. HIV AIDS, 2024, 19, 234–240 CrossRef CAS PubMed.
- Y. Li, T. Yu, H. Yan, D. Li, T. Yu, T. Yuan, A. Rahaman, S. Ali, F. Abbas, Z. Dian, X. Wu and Z. Baloch, Infect. Drug Resist., 2020, 13, 1213–1220 CrossRef CAS PubMed.
- J. M. Fettweis, M. G. Serrano, J. P. Brooks, D. J. Edwards, P. H. Girerd, H. I. Parikh, B. Huang, T. J. Arodz, L. Edupuganti, A. L. Glascock, J. Xu, N. R. Jimenez, S. C. Vivadelli, S. S. Fong, N. U. Sheth, S. Jean, V. Lee, Y. A. Bokhari, A. M. Lara, S. D. Mistry, R. A. Duckworth, S. P. Bradley, V. N. Koparde, X. V. Orenda, S. H. Milton, S. K. Rozycki, A. V. Matveyev, M. L. Wright, S. V. Huzurbazar, E. M. Jackson, E. Smirnova, J. Korlach, Y.-C. Tsai, M. R. Dickinson, J. L. Brooks, J. I. Drake, D. O. Chaffin, A. L. Sexton, M. G. Gravett, C. E. Rubens, N. R. Wijesooriya, K. D. Hendricks-Muñoz, K. K. Jefferson, J. F. Strauss and G. A. Buck, Nat. Med., 2019, 25, 1012–1021 CrossRef CAS PubMed.
- S.-Y. Teh, R. Lin, L.-H. Hung and A. P. Lee, Lab Chip, 2008, 8, 198–220 RSC.
- T. Moragues, D. Arguijo, T. Beneyton, C. Modavi, K. Simutis, A. R. Abate, J.-C. Baret, A. J. deMello, D. Densmore and A. D. Griffiths, Nat. Rev. Methods Primers, 2023, 3, 32 CrossRef CAS.
- T. Thorsen, R. W. Roberts, F. H. Arnold and S. R. Quake, Phys. Rev. Lett., 2001, 86, 4163–4166 CrossRef CAS PubMed.
- H. Song, J. D. Tice and R. F. Ismagilov, Angew. Chem., Int. Ed., 2003, 42, 768–772 CrossRef CAS PubMed.
- S. L. Anna, N. Bontoux and H. A. Stone, Appl. Phys. Lett., 2003, 82, 364–366 CrossRef CAS.
- A. Ofner, D. G. Moore, P. A. Rühs, P. Schwendimann, M. Eggersdorfer, E. Amstad, D. A. Weitz and A. R. Studart, Macromol. Chem. Phys., 2017, 218, 1600472 CrossRef.
- Z. Yu, W. Lyu, M. Yu, Q. Wang, H. Qu, R. F. Ismagilov, X. Han, D. Lai and F. Shen, Biosens. Bioelectron., 2020, 155, 112107 CrossRef CAS PubMed.
- A. R. Abate and D. A. Weitz, Lab Chip, 2011, 11, 1911–1915 RSC.
- P. Xu, C. Modavi, B. Demaree, F. Twigg, B. Liang, C. Sun, W. Zhang and A. R. Abate, Nucleic Acids Res., 2020, 48, e48 CrossRef CAS PubMed.
- R. F.-X. Tomasi, S. Sart, T. Champetier and C. N. Baroud, Cell Rep., 2020, 31, 107670 CrossRef CAS PubMed.
- J. Yun, X. Zheng, P. Xu, X. Zheng, J. Xu, C. Cao, Y. Fu, B. Xu, X. Dai, Y. Wang, H. Liu, Q. Yi, Y. Zhu, J. Wang, L. Wang, Z. Dong, L. Huang, Y. Huang and W. Du, Small, 2020, 16, 1903739 CrossRef CAS PubMed.
- A. R. Abate, T. Hung, P. Mary, J. J. Agresti and D. A. Weitz, Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 19163–19166 CrossRef CAS PubMed.
- A. Huebner, D. Bratton, G. Whyte, M. Yang, A. J. deMello, C. Abell and F. Hollfelder, Lab Chip, 2009, 9, 692–698 RSC.
- H.-D. Xi, H. Zheng, W. Guo, A. M. Gañán-Calvo, Y. Ai, C.-W. Tsao, J. Zhou, W. Li, Y. Huang, N.-T. Nguyen and S. H. Tan, Lab Chip, 2017, 17, 751–771 RSC.
- P. Zhang and A. R. Abate, Adv. Mater., 2020, 32, 2005346 CrossRef PubMed.
- A. Zinchenko, S. R. A. Devenish, B. Kintses, P.-Y. Colin, M. Fischlechner and F. Hollfelder, Anal. Chem., 2014, 86, 2526–2533 CrossRef CAS PubMed.
- M. E. Vincent, W. Liu, E. B. Haney and R. F. Ismagilov, Chem. Soc. Rev., 2010, 39, 974 RSC.
- M. Pan, F. Lyu and S. K. Y. Tang, Anal. Chem., 2015, 87, 7938–7943 CrossRef CAS PubMed.
- D.-K. Kang, M. Monsur Ali, K. Zhang, E. J. Pone and W. Zhao, TrAC, Trends Anal. Chem., 2014, 58, 145–153 CrossRef CAS.
- H. N. Joensson and H. Andersson Svahn, Angew. Chem., Int. Ed., 2012, 51, 12176–12192 CrossRef CAS PubMed.
- O. Scheler, W. Postek and P. Garstecki, Curr. Opin. Biotechnol., 2019, 55, 60–67 CrossRef CAS PubMed.
- Z. He, H. Wu, X. Yan and W. Liu, Chin. Chem. Lett., 2022, 33, 1729–1742 CrossRef CAS.
- T. S. Kaminski, O. Scheler and P. Garstecki, Lab Chip, 2016, 16, 2168–2187 RSC.
- B. Hu, P. Xu, L. Ma, D. Chen, J. Wang, X. Dai, L. Huang and W. Du, Mar. Life Sci. & Technol., 2021, 3, 169–188 Search PubMed.
- P. Liao and Y. Huang, Micromachines, 2017, 8, 231 CrossRef PubMed.
- Y. Hou, S. Chen, Y. Zheng, X. Zheng and J.-M. Lin, TrAC, Trends Anal. Chem., 2023, 158, 116897 CrossRef CAS.
- A. Stucki, J. Vallapurackal, T. R. Ward and P. S. Dittrich, Angew. Chem., Int. Ed., 2021, 60, 24368–24387 CrossRef CAS PubMed.
- A. Jain, S. Stavrakis and A. deMello, Curr. Opin. Biotechnol., 2024, 87, 103097 CrossRef CAS PubMed.
- L. Mei, M. Jin, S. Xie, Z. Yan, X. Wang, G. Zhou, A. Van Den Berg and L. Shui, Lab Chip, 2018, 18, 2806–2815 RSC.
- Z. Chen, P. Liao, F. Zhang, M. Jiang, Y. Zhu and Y. Huang, Lab Chip, 2017, 17, 235–240 RSC.
- S. Ye, C. M. Li, X. Zheng, W. H. Huang, Y. Tao, Y. H. Yu, L. M. Yang, Y. Lan, L. Ma, S. T. Bian and W. B. Du, Anal. Chem., 2022, 94, 2918–2925 CrossRef CAS PubMed.
- C. M. Li, N. Kang, S. Ye, W. H. Huang, X. Wang, C. Wang, Y. C. Li, Y. Liu, Y. Lan, L. Ma, Y. H. Zhao, Y. Han, J. Fu, D. H. Shen, L. H. Dong and W. B. Du, Adv. Sci., 2024, 11, 2309557 CrossRef CAS PubMed.
- W. Du, L. Li, K. P. Nichols and R. F. Ismagilov, Lab Chip, 2009, 9, 2286–2292 RSC.
- Z. Zhu, T. Chen, F. Huang, S. Wang, P. Zhu, R. X. Xu and T. Si, Adv. Mater., 2024, 36, 2304840 CrossRef CAS PubMed.
- F. Ahmadi, K. Samlali, P. Q. N. Vo and S. C. C. Shih, Lab Chip, 2019, 19, 524–535 RSC.
- J. Y. Tan, S. Wang, G. J. Dick, V. B. Young, D. H. Sherman, M. A. Burns and X. N. Lin, Integr. Biol., 2020, 12, 263–274 CrossRef PubMed.
- B. Hu, B. Xu, J. Yun, J. Wang, B. Xie, C. Li, Y. Yu, Y. Lan, Y. Zhu, X. Dai, Y. Huang, L. Huang, J. Pan and W. Du, Lab Chip, 2020, 20, 363–372 RSC.
- L. Mahler, M. Tovar, T. Weber, S. Brandes, M. M. Rudolph, J. Ehgartner, T. Mayr, M. T. Figge, M. Roth and E. Zang, RSC Adv., 2015, 5, 101871–101878 RSC.
- Y. Wang, M. Liu, Y. Zhang, H. Liu and L. Han, Lab Chip, 2023, 23, 1080–1096 RSC.
- S. Sart, G. Ronteix, S. Jain, G. Amselem and C. N. Baroud, Chem. Rev., 2022, 122, 7061–7096 CrossRef CAS PubMed.
- T. Orevi, S. J. Sørensen and N. Kashtan, ISME Commun., 2022, 2, 72 CrossRef PubMed.
- D. Cai, Y. Wang, J. Zou, Z. Li, E. Huang, X. Ouyang, Z. Que, Y. Luo, Z. Chen, Y. Jiang, G. Zhang, H. Wu and D. Liu, Adv. Sci., 2023, 10, 2205863 CrossRef CAS PubMed.
- B. Hu, S. Ye, D. Chen, B. Xie, R. Hu, Y. Qiao, Y. Yu, H. Yu, X. Zheng, Y. Lan and W. Du, Anal. Chem., 2021, 93, 13112–13117 CrossRef CAS PubMed.
- B. O'Donovan, D. J. Eastburn and A. R. Abate, Lab Chip, 2012, 12, 4029–4032 RSC.
- H. Yuan, Y. Pan, J. Tian, Y. Chao, J. Li and H. C. Shum, Sens. Actuators, B, 2019, 298, 126766 CrossRef CAS.
- S. S. Bithi and S. A. Vanapalli, Biomicrofluidics, 2010, 4, 044110 CrossRef PubMed.
- C. M. Ackerman, C. Myhrvold, S. G. Thakku, C. A. Freije, H. C. Metsky, D. K. Yang, S. H. Ye, C. K. Boehm, T.-S. F. Kosoko-Thoroddsen, J. Kehe, T. G. Nguyen, A. Carter, A. Kulesa, J. R. Barnes, V. G. Dugan, D. T. Hung, P. C. Blainey and P. C. Sabeti, Nature, 2020, 582, 277–282 CrossRef CAS PubMed.
- J. Kehe, A. Kulesa, A. Ortiz, C. M. Ackerman, S. G. Thakku, D. Sellers, S. Kuehn, J. Gore, J. Friedman and P. C. Blainey, Proc. Natl. Acad. Sci. U. S. A., 2019, 116, 12804–12809 CrossRef CAS PubMed.
- P. Xu, X. Zheng, Y. Tao and W. Du, Anal. Chem., 2016, 88, 3171–3177 CrossRef CAS PubMed.
- H. N. Joensson, M. Uhlén and H. A. Svahn, Lab Chip, 2011, 11, 1305–1310 RSC.
- G. C. E. Porter, S. N. F. Sikora, J. Shim, B. J. Murray and M. D. Tarn, Lab Chip, 2020, 20, 3876–3887 RSC.
- J. J. Agresti, E. Antipov, A. R. Abate, K. Ahn, A. C. Rowat, J.-C. Baret, M. Marquez, A. M. Klibanov, A. D. Griffiths and D. A. Weitz, Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 4004–4009 CrossRef CAS PubMed.
- X. Ding, S.-C. S. Lin, M. I. Lapsley, S. Li, X. Guo, C. Y. Chan, I.-K. Chiang, L. Wang, J. P. McCoy and T. J. Huang, Lab Chip, 2012, 12, 4228–4231 RSC.
- K. Zhang, Q. Liang, S. Ma, X. Mu, P. Hu, Y. Wang and G. Luo, Lab Chip, 2009, 9, 2992–2999 RSC.
- A. R. Abate, J. J. Agresti and D. A. Weitz, Appl. Phys. Lett., 2010, 96, 203509 CrossRef.
- L. Wu, P. Chen, Y. Dong, X. Feng and B.-F. Liu, Biomed. Microdevices, 2013, 15, 553–560 CrossRef CAS PubMed.
- J.-C. Baret, O. J. Miller, V. Taly, M. Ryckelynck, A. El-Harrak, L. Frenz, C. Rick, M. L. Samuels, J. B. Hutchison, J. J. Agresti, D. R. Link, D. A. Weitz and A. D. Griffiths, Lab Chip, 2009, 9, 1850–1858 RSC.
- F. Gielen, R. Hours, S. Emond, M. Fischlechner, U. Schell and F. Hollfelder, Proc. Natl. Acad. Sci. U. S. A., 2016, 113, E7383–E7389 CrossRef CAS PubMed.
- E. J. Medcalf, M. Gantz, T. S. Kaminski and F. Hollfelder, Anal. Chem., 2023, 95, 4597–4604 CrossRef CAS PubMed.
- W. J. Watterson, M. Tanyeri, A. R. Watson, C. M. Cham, Y. Shan, E. B. Chang, A. M. Eren and S. Tay, eLife, 2020, 9, e56998 CrossRef CAS PubMed.
- A. Gérard, A. Woolfe, G. Mottet, M. Reichen, C. Castrillon, V. Menrath, S. Ellouze, A. Poitou, R. Doineau, L. Briseno-Roa, P. Canales-Herrerias, P. Mary, G. Rose, C. Ortega, M. Delincé, S. Essono, B. Jia, B. Iannascoli, O. Richard-Le Goff, R. Kumar, S. N. Stewart, Y. Pousse, B. Shen, K. Grosselin, B. Saudemont, A. Sautel-Caillé, A. Godina, S. McNamara, K. Eyer, G. A. Millot, J. Baudry, P. England, C. Nizak, A. Jensen, A. D. Griffiths, P. Bruhns and C. Brenan, Nat. Biotechnol., 2020, 38, 715–721 CrossRef PubMed.
- E. K. Bowman and H. S. Alper, Trends Biotechnol., 2020, 38, 701–714 CrossRef CAS PubMed.
- R. Obexer, A. Godina, X. Garrabou, P. R. E. Mittl, D. Baker, A. D. Griffiths and D. Hilvert, Nat. Chem., 2017, 9, 50–56 CrossRef CAS PubMed.
- D. A. Holland-Moritz, M. K. Wismer, B. F. Mann, I. Farasat, P. Devine, E. D. Guetschow, I. Mangion, C. J. Welch, J. C. Moore, S. Sun and R. T. Kennedy, Angew. Chem., Int. Ed., 2020, 59, 4470–4477 CrossRef CAS PubMed.
- Y. Xie, Z. Chen, D. Cai, D. Huang, E. Huang, X. Yang, T. Zhang, H. Wen, Y. Wang, M. Zhao, D. Liu and B. Xu, Anal. Chem., 2024, 96, 12561–12569 CAS.
- T. Henkel, G. Mayer, J. Hampl, J. Cao, L. Ehrhardt, A. Schober and G. Groß, Micromachines, 2022, 13, 1022 CrossRef PubMed.
- D. Josephides, S. Davoli, W. Whitley, R. Ruis, R. Salter, S. Gokkaya, M. Vallet, D. Matthews, G. Benazzi, E. Shvets, F. Gesellchen, D. Geere, X. Liu, X. Li, B. Mackworth, W. Young, Z. Owen, C. Smith, D. Starkie, J. White, B. Sweeney, M. Hinchliffe, S. Tickle, D. J. Lightwood, M. Rehak, F. F. Craig and D. Holmes, SLAS Technol., 2020, 25, 177–189 CrossRef PubMed.
- C. Zong, S. Lu, A. R. Chapman and X. S. Xie, Science, 2012, 338, 1622–1626 CrossRef CAS PubMed.
- W. Zheng, S. Zhao, Y. Yin, H. Zhang, D. M. Needham, E. D. Evans, C. L. Dai, P. J. Lu, E. J. Alm and D. A. Weitz, Science, 2022, 376, eabm1483 CrossRef CAS PubMed.
- P. Ma, H. M. Amemiya, L. L. He, S. J. Gandhi, R. Nicol, R. P. Bhattacharyya, C. S. Smillie and D. T. Hung, Cell, 2023, 186, 877–891.e14 CrossRef CAS PubMed.
- K. Arikawa, K. Ide, M. Kogawa, T. Saeki, T. Yoda, T. Endoh, A. Matsuhashi, H. Takeyama and M. Hosokawa, Microbiome, 2021, 9, 202 CrossRef CAS PubMed.
- H. Telenius, N. P. Carter, C. E. Bebb, M. Nordenskjöld, B. A. J. Ponder and A. Tunnacliffe, Genomics, 1992, 13, 718–725 CrossRef CAS PubMed.
- F. B. Dean, S. Hosono, L. Fang, X. Wu, A. F. Faruqi, P. Bray-Ward, Z. Sun, Q. Zong, Y. Du, J. Du, M. Driscoll, W. Song, S. F. Kingsmore, M. Egholm and R. S. Lasken, Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 5261–5266 CrossRef CAS PubMed.
- C. Chen, D. Xing, L. Tan, H. Li, G. Zhou, L. Huang and X. S. Xie, Science, 2017, 356, 189–194 CrossRef CAS PubMed.
- V. Gonzalez-Pena, S. Natarajan, Y. Xia, D. Klein, R. Carter, Y. Pang, B. Shaner, K. Annu, D. Putnam, W. Chen, J. Connelly, S. Pruett-Miller, X. Chen, J. Easton and C. Gawad, Proc. Natl. Acad. Sci. U. S. A., 2021, 118, e2024176118 CrossRef CAS PubMed.
- L. Wen and F. Tang, Precis. Clin. Med., 2022, 5, pbac002 CrossRef PubMed.
- D. Ramsköld, S. Luo, Y.-C. Wang, R. Li, Q. Deng, O. R. Faridani, G. A. Daniels, I. Khrebtukova, J. F. Loring, L. C. Laurent, G. P. Schroth and R. Sandberg, Nat. Biotechnol., 2012, 30, 777–782 CrossRef PubMed.
- A. J. Reid, A. M. Talman, H. M. Bennett, A. R. Gomes, M. J. Sanders, C. J. R. Illingworth, O. Billker, M. Berriman and M. K. Lawniczak, eLife, 2018, 7, e33105 CrossRef PubMed.
- T. Hashimshony, F. Wagner, N. Sher and I. Yanai, Cell Rep., 2012, 2, 666–673 CrossRef CAS PubMed.
- C. S. McGinnis, D. M. Patterson, J. Winkler, D. N. Conrad, M. Y. Hein, V. Srivastava, J. L. Hu, L. M. Murrow, J. S. Weissman, Z. Werb, E. D. Chow and Z. J. Gartner, Nat. Methods, 2019, 16, 619–626 CrossRef CAS PubMed.
- C. Rinke, P. Schwientek, A. Sczyrba, N. N. Ivanova, I. J. Anderson, J.-F. Cheng, A. Darling, S. Malfatti, B. K. Swan, E. A. Gies, J. A. Dodsworth, B. P. Hedlund, G. Tsiamis, S. M. Sievert, W.-T. Liu, J. A. Eisen, S. J. Hallam, N. C. Kyrpides, R. Stepanauskas, E. M. Rubin, P. Hugenholtz and T. Woyke, Nature, 2013, 499, 431–437 CrossRef CAS PubMed.
- Z. Diao, L. Kan, Y. Zhao, H. Yang, J. Song, C. Wang, Y. Liu, F. Zhang, T. Xu, R. Chen, Y. Ji, X. Wang, X. Jing, J. Xu, Y. Li and B. Ma, mLife, 2022, 1, 448–459 CrossRef CAS PubMed.
- T. Xu, Y. Li, X. Han, L. Kan, J. Ren, L. Sun, Z. Diao, Y. Ji, P. Zhu, J. Xu and B. Ma, Lab Chip, 2023, 23, 125–135 RSC.
- Y. Kang, I. McMillan, M. H. Norris and T. T. Hoang, Nat. Protoc., 2015, 10, 974–984 CrossRef CAS PubMed.
- M. R. Emmert-Buck, R. F. Bonner, P. D. Smith, R. F. Chuaqui, Z. Zhuang, S. R. Goldstein, R. A. Weiss and L. A. Liotta, Science, 1996, 274, 998–1001 CrossRef CAS PubMed.
- S. B. Blattman, W. Jiang, P. Oikonomou and S. Tavazoie, Nat. Microbiol., 2020, 5, 1192–1201 CrossRef CAS PubMed.
- A. Kuchina, L. M. Brettner, L. Paleologu, C. M. Roco, A. B. Rosenberg, A. Carignano, R. Kibler, M. Hirano, R. W. DePaolo and G. Seelig, Science, 2021, 371, eaba5257 CrossRef CAS PubMed.
- A. Pryszlak, T. Wenzel, K. W. Seitz, F. Hildebrand, E. Kartal, M. R. Cosenza, V. Benes, P. Bork and C. A. Merten, Cells Rep. Methods, 2022, 2, 100137 CrossRef CAS PubMed.
- R. Chijiiwa, M. Hosokawa, M. Kogawa, Y. Nishikawa, K. Ide, C. Sakanashi, K. Takahashi and H. Takeyama, Microbiome, 2020, 8, 5 CrossRef CAS PubMed.
- H. Aoki, Y. Masahiro, M. Shimizu, Y. Hongoh, M. Ohkuma and Y. Yamagata, Sci. Rep., 2022, 12, 17014 CrossRef CAS PubMed.
- R. Zilionis, J. Nainys, A. Veres, V. Savova, D. Zemmour, A. M. Klein and L. Mazutis, Nat. Protoc., 2017, 12, 44–73 CrossRef CAS PubMed.
- E. Z. Macosko, A. Basu, R. Satija, J. Nemesh, K. Shekhar, M. Goldman, I. Tirosh, A. R. Bialas, N. Kamitaki, E. M. Martersteck, J. J. Trombetta, D. A. Weitz, J. R. Sanes, A. K. Shalek, A. Regev and S. A. McCarroll, Cell, 2015, 161, 1202–1214 CrossRef CAS PubMed.
- Y. Fu, C. Li, S. Lu, W. Zhou, F. Tang, X. S. Xie and Y. Huang, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, 11923–11928 CrossRef CAS PubMed.
- A. M. Sidore, F. Lan, S. W. Lim and A. R. Abate, Nucleic Acids Res., 2016, 44, e66 CrossRef PubMed.
- F. Lan, B. Demaree, N. Ahmed and A. R. Abate, Nat. Biotechnol., 2017, 35, 640–646 CrossRef CAS PubMed.
- M. Kogawa, M. Hosokawa, Y. Nishikawa, K. Mori and H. Takeyama, Sci. Rep., 2018, 8, 2059 CrossRef PubMed.
- D. R. Mende, F. O. Aylward, J. M. Eppley, T. N. Nielsen and E. F. DeLong, Front. Microbiol., 2016, 7, 143 Search PubMed.
- F. Lan, J. Saba, T. D. Ross, Z. Zhou, K. Krauska, K. Anantharaman, R. Landick and O. S. Venturelli, Nat. Methods, 2024, 21, 228–235 CrossRef CAS PubMed.
- C. Sun, L. Liu, L. Pérez, X. Li, Y. Liu, P. Xu, E. A. Boritz, J. I. Mullins and A. R. Abate, Nat. Biomed. Eng., 2022, 6, 1004–1012 CrossRef CAS PubMed.
- E. A. Franzosa, T. Hsu, A. Sirota-Madi, A. Shafquat, G. Abu-Ali, X. C. Morgan and C. Huttenhower, Nat. Rev. Microbiol., 2015, 13, 360–372 CrossRef CAS PubMed.
- G. Avital, R. Avraham, A. Fan, T. Hashimshony, D. T. Hung and I. Yanai, Genome Biol., 2017, 18, 200 CrossRef PubMed.
- Z. Xu, Y. Wang, K. Sheng, R. Rosenthal, N. Liu, X. Hua, T. Zhang, J. Chen, M. Song, Y. Lv, S. Zhang, Y. Huang, Z. Wang, T. Cao, Y. Shen, Y. Jiang, Y. Yu, Y. Chen, G. Guo, P. Yin, D. A. Weitz and Y. Wang, Nat. Commun., 2023, 14, 5130 CrossRef CAS PubMed.
- R. McNulty, D. Sritharan, S. H. Pahng, J. P. Meisch, S. Liu, M. A. Brennan, G. Saxer, S. Hormoz and A. Z. Rosenthal, Nat. Microbiol., 2023, 8, 934–945 CrossRef CAS PubMed.
- H. Meng, T. Zhang, Z. Wang, Y. Zhu, Y. Yu, H. Chen, J. Chen, F. Wang, Y. Yu, X. Hua and Y. Wang, Angew. Chem., Int. Ed., 2024, 63, e202400538 CrossRef CAS PubMed.
- K. Karagiannis, S. Gannavaram, C. Verma, T. Pacheco-Fernandez, P. Bhattacharya, H. L. Nakhasi and A. R. Satoskar, Cell Rep., 2023, 42, 113097 CrossRef CAS PubMed.
- K. Ratnasiri, A. J. Wilk, M. J. Lee, P. Khatri and C. A. Blish, Semin. Immunopathol., 2023, 45, 71–89 CrossRef CAS PubMed.
- A. Almeida, S. Nayfach, M. Boland, F. Strozzi, M. Beracochea, Z. J. Shi, K. S. Pollard, E. Sakharova, D. H. Parks, P. Hugenholtz, N. Segata, N. C. Kyrpides and R. D. Finn, Nat. Biotechnol., 2021, 39, 105–114 CrossRef CAS PubMed.
- C. Liu, M.-X. Du, R. Abuduaini, H.-Y. Yu, D.-H. Li, Y.-J. Wang, N. Zhou, M.-Z. Jiang, P.-X. Niu, S.-S. Han, H.-H. Chen, W.-Y. Shi, L. Wu, Y.-H. Xin, J. Ma, Y. Zhou, C.-Y. Jiang, H.-W. Liu and S.-J. Liu, Microbiome, 2021, 9, 119 CrossRef CAS PubMed.
- A. Diakite, G. Dubourg and D. Raoult, Microb. Pathog., 2021, 150, 104698 CrossRef PubMed.
- C. H. J. Schmitz, A. C. Rowat, S. Köster and D. A. Weitz, Lab Chip, 2009, 9, 44–49 RSC.
- J.-G. Xu, M.-S. Huang, H.-F. Wang and Q. Fang, Anal. Chem., 2019, 91, 10757–10763 CrossRef CAS PubMed.
- C.-Y. Jiang, L. Dong, J.-K. Zhao, X. Hu, C. Shen, Y. Qiao, X. Zhang, Y. Wang, R. F. Ismagilov, S.-J. Liu and W. Du, Appl. Environ. Microbiol., 2016, 82, 2210–2218 CrossRef CAS PubMed.
- A. L. McCully, M. Loop Yao, K. K. Brower, P. M. Fordyce and A. M. Spormann, ISME Commun., 2023, 3, 47 CrossRef PubMed.
- J. Park, A. Kerner, M. A. Burns and X. N. Lin, PLoS One, 2011, 6, e17019 CrossRef CAS PubMed.
- D.-K. Kang, X. Gong, S. Cho, J. Kim, J. B. Edel, S.-I. Chang, J. Choo and A. J. deMello, Anal. Chem., 2015, 87, 10770–10778 CrossRef CAS PubMed.
- A. Afrizal, T. C. A. Hitch, A. Viehof, N. Treichel, T. Riedel, B. Abt, E. M. Buhl, D. Kohlheyer, J. Overmann and T. Clavel, Environ. Microbiol., 2022, 24, 3861–3881 CrossRef CAS PubMed.
- L. Ma, J. Kim, R. Hatzenpichler, M. A. Karymov, N. Hubert, I. M. Hanan, E. B. Chang and R. F. Ismagilov, Proc. Natl. Acad. Sci. U. S. A., 2014, 111, 9768–9773 CrossRef CAS PubMed.
- B. Xie, J. Wang, Y. Nie, J. Tian, Z. Wang, D. Chen, B. Hu, X.-L. Wu and W. Du, Proc. Natl. Acad. Sci. U. S. A., 2022, 119, e2215990119 CrossRef CAS PubMed.
- D. K. W. Man, S. M. Hermans, M. Taubert, S. L. Garcia, S. Hengoju, K. Küsel and M. A. Rosenbaum, ISME Commun., 2024, 4, ycae080 CrossRef PubMed.
- N. J. Reichart, Z. J. Jay, V. Krukenberg, A. E. Parker, R. L. Spietz and R. Hatzenpichler, ISME J., 2020, 14, 2851–2861 CrossRef CAS PubMed.
- X. Wang, L. Ren, Y. Su, Y. Ji, Y. Liu, C. Li, X. Li, Y. Zhang, W. Wang, Q. Hu, D. Han, J. Xu and B. Ma, Anal. Chem., 2017, 89, 12569–12577 CrossRef CAS PubMed.
- G. Batani, K. Bayer, J. Böge, U. Hentschel and T. Thomas, Sci. Rep., 2019, 9, 18618 CrossRef CAS PubMed.
- T. Xu, Y. Gong, X. Su, P. Zhu, J. Dai, J. Xu and B. Ma, Small, 2020, 16, 2001172 CrossRef CAS PubMed.
- F. Hu, L. Shi and W. Min, Nat. Methods, 2019, 16, 830–842 CrossRef CAS PubMed.
- Y. Wang, Z. Chen, P. Zeng, A. Cao, T. Zhang and Y. Li, J. Mater. Chem. C, 2023, 11, 4486–4491 RSC.
- S. J. Spencer, M. V. Tamminen, S. P. Preheim, M. T. Guo, A. W. Briggs, I. L. Brito, D. A. Weitz, L. K. Pitkänen, F. Vigneault, M. P. Virta and E. J. Alm, ISME J., 2016, 10, 427–436 CrossRef CAS PubMed.
- E. G. Sakowski, K. Arora-Williams, F. Tian, A. A. Zayed, O. Zablocki, M. B. Sullivan and S. P. Preheim, Nat. Microbiol., 2021, 6, 630–642 CrossRef CAS PubMed.
- W. Shen, D. Wang, J. Li, Y. Liu, Y. Wang, X. Yang, X. Peng, B. Xie, L. Su, Z. Wei, Q. He, Z. Wang, K. Feng, W. Du and Y. Deng, mLife, 2025 DOI:10.1002/mlf2.12159.
- K. L. Cross, J. H. Campbell, M. Balachandran, A. G. Campbell, C. J. Cooper, A. Griffen, M. Heaton, S. Joshi, D. Klingeman, E. Leys, Z. Yang, J. M. Parks and M. Podar, Nat. Biotechnol., 2019, 37, 1314–1321 CrossRef CAS PubMed.
- M. Hosokawa, N. Iwai, K. Arikawa, T. Saeki, T. Endoh, K. Kamata, T. Yoda, S. Tsuda and H. Takeyama, J. Biosci. Bioeng., 2023, 136, 58–66 CrossRef CAS PubMed.
- P. T. Kumar, D. Decrop, S. Safdar, I. Passaris, T. Kokalj, R. Puers, A. Aertsen, D. Spasic and J. Lammertyn, Micromachines, 2020, 11, 308 CrossRef PubMed.
- S. Jakiela, T. S. Kaminski, O. Cybulski, D. B. Weibel and P. Garstecki, Angew. Chem., Int. Ed., 2013, 52, 8908–8911 CrossRef CAS PubMed.
- D. Taylor, N. Verdon, P. Lomax, R. J. Allen and S. Titmuss, Phys. Biol., 2022, 19, 026003 CrossRef CAS PubMed.
- S. Mellouli, B. Monterroso, H. R. Vutukuri, E. te Brinke, V. Chokkalingam, G. Rivas and W. T. Huck, Soft Matter, 2013, 9, 10493–10500 RSC.
- E. C. Carnes, D. M. Lopez, N. P. Donegan, A. Cheung, H. Gresham, G. S. Timmins and C. J. Brinker, Nat. Chem. Biol., 2010, 6, 41–45 CrossRef CAS PubMed.
- Z. Jin, M. Nie, R. Hu, T. Zhao, J. Xu, D. Chen, J. Yun, L. Z. Ma and W. Du, Small, 2018, 14, 1800658 CrossRef PubMed.
- J. Luo, J. Li, H. Yang, J. Yu and H. Wei, J. Clin. Microbiol., 2017, 55, 2946–2955 CrossRef CAS PubMed.
- D.-K. Kang, M. M. Ali, K. Zhang, S. S. Huang, E. Peterson, M. A. Digman, E. Gratton and W. Zhao, Nat. Commun., 2014, 5, 5427 CrossRef CAS PubMed.
- A. M. Kaushik, K. Hsieh, L. Chen, D. J. Shin, J. C. Liao and T.-H. Wang, Biosens. Bioelectron., 2017, 97, 260–266 CrossRef CAS PubMed.
- D. Yang, Z. Yu, M. Zheng, W. Yang, Z. Liu, J. Zhou and L. Huang, Lab Chip, 2023, 23, 3961–3977 RSC.
- R. H. Hsu, R. L. Clark, J. W. Tan, J. C. Ahn, S. Gupta, P. A. Romero and O. S. Venturelli, Cell Syst., 2019, 9, 229–242.e4 CrossRef CAS PubMed.
- Y. Fan and O. Pedersen, Nat. Rev. Microbiol., 2021, 19, 55–71 CrossRef CAS PubMed.
- S. Sharma, P. Hegde, S. Panda, M. O. Orimoloye and C. C. Aldrich, Curr. Opin. Microbiol., 2023, 71, 102234 CrossRef CAS PubMed.
- S. Siedler, N. K. Khatri, A. Zsohár, I. Kjærbølling, M. Vogt, P. Hammar, C. F. Nielsen, J. Marienhagen, M. O. A. Sommer and H. N. Joensson, ACS Synth. Biol., 2017, 6, 1860–1869 CrossRef PubMed.
- J. Abatemarco, M. F. Sarhan, J. M. Wagner, J.-L. Lin, L. Liu, W. Hassouneh, S.-F. Yuan, H. S. Alper and A. R. Abate, Nat. Commun., 2017, 8, 332 CrossRef PubMed.
- L. Mahler, S. P. Niehs, K. Martin, T. Weber, K. Scherlach, C. Hertweck, M. Roth and M. A. Rosenbaum, eLife, 2021, 10, e64774 CrossRef CAS PubMed.
- K. Samlali, C. L. Alves, M. Jezernik and S. C. C. Shih, Microsyst. Nanoeng., 2022, 8, 123 CrossRef CAS PubMed.
- J. M. Wagner, L. Liu, S.-F. Yuan, M. V. Venkataraman, A. R. Abate and H. S. Alper, Metab. Eng., 2018, 47, 346–356 CrossRef CAS PubMed.
- A. Das, C. Weise, M. Polack, R. D. Urban, B. Krafft, S. Hasan, H. Westphal, R. Warias, S. Schmidt, T. Gulder and D. Belder, J. Am. Chem. Soc., 2022, 144, 10353–10360 CrossRef CAS PubMed.
- K. Wink, M. Van Der Loh, N. Hartner, M. Polack, C. Dusny, A. Schmid and D. Belder, Angew. Chem., Int. Ed., 2022, 61, e202204098 CrossRef CAS PubMed.
- L. Mahler, K. Wink, R. J. Beulig, K. Scherlach, M. Tovar, E. Zang, K. Martin, C. Hertweck, D. Belder and M. Roth, Sci. Rep., 2018, 8, 13087 CrossRef PubMed.
- K. Wink, L. Mahler, J. R. Beulig, S. K. Piendl, M. Roth and D. Belder, Anal. Bioanal. Chem., 2018, 410, 7679–7687 CrossRef CAS PubMed.
- C. A. Smith, X. Li, T. H. Mize, T. D. Sharpe, E. I. Graziani, C. Abell and W. T. S. Huck, Anal. Chem., 2013, 85, 3812–3816 CrossRef CAS PubMed.
- C. Kim, M. Holm, I. Frost, M. Hasso-Agopsowicz and K. Abbas, BMJ Global Health, 2023, 8, e011341 CrossRef PubMed.
- R. Yang, A. Paparini, P. Monis and U. Ryan, Int. J. Parasitol., 2014, 44, 1105–1113 CrossRef CAS PubMed.
- C. Tan, D. Fan, N. Wang, F. Wang, B. Wang, L. Zhu and Y. Guo, View, 2021, 2, 20200082 CrossRef CAS PubMed.
- K. Hsieh, K. E. Mach, P. Zhang, J. C. Liao and T.-H. Wang, Acc. Chem. Res., 2022, 55, 123–133 CrossRef CAS PubMed.
- N. G. Schoepp, E. M. Khorosheva, T. S. Schlappi, M. S. Curtis, R. M. Humphries, J. A. Hindler and R. F. Ismagilov, Angew. Chem., Int. Ed., 2016, 55, 9557–9561 CrossRef CAS PubMed.
- N. G. Schoepp, T. S. Schlappi, M. S. Curtis, S. S. Butkovich, S. Miller, R. M. Humphries and R. F. Ismagilov, Sci. Transl. Med., 2017, 9, eaal3693 CrossRef PubMed.
- A. Ruszczak, S. Bartkova, M. Zapotoczna, O. Scheler and P. Garstecki, Curr. Opin. Biotechnol., 2022, 76, 102755 CrossRef CAS PubMed.
- Y. Wang, D. Cai, X. Ouyang, H. He, Y. Liu, J. Zou, Z. Chen, B. Wu, H. Wu and D. Liu, Biosens. Bioelectron., 2023, 220, 114863 CrossRef CAS PubMed.
- X. Li, X. Liu, Z. Yu, Y. Luo, Q. Hu, Z. Xu, J. Dai, N. Wu and F. Shen, Lab Chip, 2022, 22, 3952–3960 RSC.
- J. Riti, G. Sutra, T. Naas, H. Volland, S. Simon and K. Perez-Toralla, Biosens. Bioelectron., 2024, 257, 116301 CrossRef CAS PubMed.
- K. Faust and J. Raes, F1000Research, 2016, 5, 1519 Search PubMed.
- S. Nagpal, R. Singh, D. Yadav and S. S. Mande, Nucleic Acids Res., 2020, 48, W572–W579 CrossRef CAS PubMed.
- S. Peschel, C. L. Müller, E. Von Mutius, A.-L. Boulesteix and M. Depner, Briefings Bioinf., 2021, 22, bbaa290 CrossRef PubMed.
- C. Vitalis and T. Wenzel, Curr. Opin. Biotechnol., 2023, 82, 102966 CrossRef CAS PubMed.
- K. Leung, H. Zahn, T. Leaver, K. M. Konwar, N. W. Hanson, A. P. Pagé, C.-C. Lo, P. S. Chain, S. J. Hallam and C. L. Hansen, Proc. Natl. Acad. Sci. U. S. A., 2012, 109, 7665–7670 CrossRef CAS PubMed.
- J. Q. Yu, W. Huang, L. K. Chin, L. Lei, Z. P. Lin, W. Ser, H. Chen, T. C. Ayi, P. H. Yap, C. H. Chen and A. Q. Liu, Lab Chip, 2014, 14, 3519–3524 RSC.
- L. J. Barkal, A. B. Theberge, C.-J. Guo, J. Spraker, L. Rappert, J. Berthier, K. A. Brakke, C. C. C. Wang, D. J. Beebe, N. P. Keller and E. Berthier, Nat. Commun., 2016, 7, 10610 CrossRef CAS PubMed.
- M. Schwarzer, A. Repa, C. Daniel, I. Schabussova, T. Hrncir, B. Pot, R. Stepankova, T. Hudcovic, A. Pollak and H. Tlaskalova-Hogenova, Allergy, 2011, 66, 368–375 CrossRef CAS PubMed.
- K. Fu, A. H. K. Cheung, C. C. Wong, W. Liu, Y. Zhou, F. Wang, P. Huang, K. Yuan, O. O. Coker, Y. Pan, D. Chen, N. M. Lam, M. Gao, X. Zhang, H. Huang, K. F. To, J. J. Y. Sung and J. Yu, Cell, 2024, 187, 882–896.e17 CrossRef CAS PubMed.
- J. Zhang, S. Wan, H. Zhou, J. Du, Y. Li, H. Zhu, L. Weng, X. Ding and L. Wang, ACS Nano, 2024, 18, 9613–9626 CrossRef CAS PubMed.
- X. Yang, W. Nie, C. Wang, Z. Fang and L. Shang, Biomaterials, 2024, 308, 122564 CrossRef CAS PubMed.
- X. Xie, Q. Li, L. Jia, H. Yuan, T. Guo and T. Meng, Adv. Healthcare Mater., 2023, 12, 2202954 CrossRef CAS PubMed.
- Y. Yin, Z. Li, H. Gao, D. Zhou, Z. Zhu, L. Tao, W. Guan, Y. Gao, Y. Song and M. Wang, Nano Lett., 2024, 24, 1081–1089 CrossRef CAS PubMed.
- S. I. Lee, Y. Y. Choi, S. G. Kang, T. H. Kim, J. W. Choi, Y. J. Kim, T.-H. Kim, T. Kang and B. G. Chung, Polymers, 2022, 14, 3752 CrossRef CAS PubMed.
- B. Li, L. Zhang, Y. Yin, A. Chen, B. R. Seo, J. Lou, D. J. Mooney and D. A. Weitz, Matter, 2024, 7, 3447–3468 CrossRef CAS PubMed.
- D. E. Ingber, Nat. Rev. Genet., 2022, 23, 467–491 CrossRef CAS PubMed.
- J. D. Caplin, N. G. Granados, M. R. James, R. Montazami and N. Hashemi, Adv. Healthcare Mater., 2015, 4, 1426–1450 CrossRef CAS PubMed.
- J. Barrila, A. Crabbé, J. Yang, K. Franco, S. D. Nydam, R. J. Forsyth, R. R. Davis, S. Gangaraju, C. M. Ott, C. B. Coyne, M. J. Bissell and C. A. Nickerson, Infect. Immun., 2018, 86, e00282-18 CrossRef PubMed.
- Y. Lee, M.-H. Kim, D. R. Alves, S. Kim, L. P. Lee, J. H. Sung and S. Park, Toxins, 2021, 13, 775 CrossRef CAS PubMed.
- J. Yang, Y. Hirai, K. Iida, S. Ito, M. Trumm, S. Terada, R. Sakai, T. Tsuchiya, O. Tabata and K. Kamei, Commun. Biol., 2023, 6, 310 CrossRef CAS PubMed.
- R. Kim and J. H. Sung, Adv. Healthcare Mater., 2024, 13, 2302777 CrossRef CAS PubMed.
- Y.-C. Chen, Y.-H. Cheng, H. S. Kim, P. N. Ingram, J. E. Nor and E. Yoon, Lab Chip, 2014, 14, 2941–2947 RSC.
- A. Lashkaripour, D. P. McIntyre, S. G. K. Calhoun, K. Krauth, D. M. Densmore and P. M. Fordyce, Nat. Commun., 2024, 15, 83 CrossRef CAS PubMed.
- C. Barnes, A. R. Sonwane, E. C. Sonnenschein and F. Del Giudice, Phys. Fluids, 2023, 35, 092003 CrossRef CAS.
- M. Sesen and G. Whyte, Sci. Rep., 2020, 10, 8736 CrossRef CAS PubMed.
- S. Yu, X. Li, W. Lu, H. Li, Y. V. Fu and F. Liu, Anal. Chem., 2021, 93, 11089–11098 CrossRef CAS PubMed.
- X. Wang, L. Ren, Z. Diao, Y. He, J. Zhang, M. Liu, Y. Li, L. Sun, R. Chen, Y. Ji, J. Xu and B. Ma, Adv. Sci., 2023, 10, 2207497 CrossRef CAS PubMed.
- M. A. Oberhardt, R. Zarecki, S. Gronow, E. Lang, H.-P. Klenk, U. Gophna and E. Ruppin, Nat. Commun., 2015, 6, 8493 CrossRef CAS PubMed.
- J. Koblitz, P. Halama, S. Spring, V. Thiel, C. Baschien, R. L. Hahnke, M. Pester, J. Overmann and L. C. Reimer, Nucleic Acids Res., 2023, 51, D1531–D1538 CrossRef PubMed.
- T. Hashizume and B.-W. Ying, Biotechnol. Adv., 2024, 70, 108293 CrossRef CAS PubMed.
- Y. Huang, R. U. Sheth, S. Zhao, L. A. Cohen, K. Dabaghi, T. Moody, Y. Sun, D. Ricaurte, M. Richardson, F. Velez-Cortes, T. Blazejewski, A. Kaufman, C. Ronda and H. H. Wang, Nat. Biotechnol., 2023, 41, 1424–1433 CrossRef CAS PubMed.
- E. Muller, I. Shiryan and E. Borenstein, Nat. Commun., 2024, 15, 2621 CrossRef CAS PubMed.
- A. M. Eren and J. F. Banfield, Cell, 2024, 187, 5151–5170 CrossRef CAS PubMed.
- V. Lloréns-Rico, J. A. Simcock, G. R. B. Huys and J. Raes, Cell, 2022, 185, 2725–2738 CrossRef PubMed.
- M. T. Sorbara and E. G. Pamer, Nat. Rev. Microbiol., 2022, 20, 365–380 CrossRef CAS PubMed.
- Z. Diao, X. Wang, J. Zhang, A. Ge, T. Xu, L. Kan, Y. Li, Y. Ji, X. Jing, J. Xu and B. Ma, Biosens. Bioelectron., 2023, 240, 115639 CrossRef CAS PubMed.
Footnote |
† These authors contributed equally to this work. |
|
This journal is © The Royal Society of Chemistry 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.